1
|
Mukherjee D, Satyavolu S, Cioffi S, Thomas A, Li Y, Nayak L. Protocols for circulating neutrophil depletion in neonatal C57Bl/6 mice. J Leukoc Biol 2025; 117:qiaf005. [PMID: 39821323 DOI: 10.1093/jleuko/qiaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025] Open
Abstract
Murine neonatal neutrophil depletion strategies have problems achieving deep neutrophil clearance and accurate residual neutrophil fraction detection. An isotype switch method can achieve profound neutrophil clearance using a combination of anti-Ly6G and antirat κ Ig light chain antibodies in adult C57Bl/6 mice, proven by extra- and intracellular Ly6G detection by flow cytometry. We adapted this technique to neonatal mice, testing four neutrophil depletion strategies in the peripheral circulation, bone marrow, and spleen. Four protocols were tested: P3 Ly6G and P1-3 Ly6G (anti-Ly6G on postnatal days [P] 3 and 1-3, respectively), and P3 Dual and P1-3 Dual (anti-Ly6G and antirat κ Ig light chain on P3 and P1-3, respectively). Intracellular and extracellular Ly6G presence was detected using flow cytometry. Isotype control antibodies were used as controls. P1-3 Dual protocol achieved significantly better neutrophil depletion than the P1-3 Ly6G or P3 Ly6G protocols (97% vs 74% and 97% vs 50%, respectively) in the peripheral circulation. The P3 Dual protocol alone was enough to achieve significantly better neutrophil clearance (93%) than any of the Ly6G alone protocols. The Ly6G alone protocols led to near-total elimination of extracellular Ly6G. However, there was a significant presence of intracellular Ly6G in the CD45+ cell population, evading detection by extracellular Ly6G antibody-based detection methods. P3 protocols perform better than P1-3 protocols for bone marrow and splenic neutrophil clearance. Thus, the P3 Dual protocol might be the most effective and ethical protocol to induce profound neutrophil depletion in neonatal mice, an alternative to daily anti-Ly6G injections.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Department of Pediatrics, Case Western Reserve University School of Medicine, 11100 Euclid Ave, Cleveland, OH 44106, United States
- Rainbow Babies and Children's Hospital, 11100 Euclid Ave, Cleveland, OH 44106, United States
| | - Sriram Satyavolu
- The Ohio State University College of Medicine, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Sarah Cioffi
- Department of Pediatrics, Case Western Reserve University School of Medicine, 11100 Euclid Ave, Cleveland, OH 44106, United States
| | - Asha Thomas
- Case Comprehensive Cancer Center, 2103 Cornell Rd, Cleveland, OH 44106, United States
| | - Yuexin Li
- Case Comprehensive Cancer Center, 2103 Cornell Rd, Cleveland, OH 44106, United States
| | - Lalitha Nayak
- Case Comprehensive Cancer Center, 2103 Cornell Rd, Cleveland, OH 44106, United States
- Division of Hematology-Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States
| |
Collapse
|
2
|
Whitaker R, Sung S, Tylek T, Risser GE, O'Brien EM, Chua PE, Li T, Petrie RJ, Han L, Binder-Markey BI, Spiller KL. Effects of injury size on local and systemic immune cell dynamics in volumetric muscle loss. NPJ Regen Med 2025; 10:9. [PMID: 39939310 PMCID: PMC11822203 DOI: 10.1038/s41536-025-00397-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
We took a systems approach to the analysis of macrophage phenotype in regenerative and fibrotic volumetric muscle loss outcomes in mice together with analysis of systemic inflammation and of other leukocytes in the muscle, spleen, and bone marrow. Differences in expression of macrophage phenotype markers occurred as early as day 1, persisted to at least day 28, and were associated with increased numbers of leukocytes in the muscle and bone marrow, increased pro-inflammatory marker expression in splenic macrophages, and changes in the levels of pro-inflammatory cytokines in the blood. The most prominent differences were in muscle neutrophils, which were much more abundant in fibrotic outcomes compared to regenerative outcomes at day 1 after injury. However, neutrophil depletion had little to no effect on macrophage phenotype or on muscle repair outcomes. Together, these results suggest that the entire system of immune cell interactions must be considered to improve muscle repair outcomes.
Collapse
Affiliation(s)
- Ricardo Whitaker
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Samuel Sung
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Tina Tylek
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Gregory E Risser
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Erin M O'Brien
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Phoebe Ellin Chua
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Thomas Li
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Ryan J Petrie
- Department of Biology, College of Arts & Sciences, Drexel University, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Benjamin I Binder-Markey
- Department of Physical Therapy & Rehabilitation Sciences, College of Nursing and Health Professions, Drexel University, Philadelphia, PA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Tan H, Jiang Y, Shen L, Nuerhashi G, Wen C, Gu L, Wang Y, Qi H, Cao F, Huang T, Liu Y, Xie W, Deng W, Fan W. Cryoablation-induced neutrophil Ca 2+ elevation and NET formation exacerbate immune escape in colorectal cancer liver metastasis. J Exp Clin Cancer Res 2024; 43:319. [PMID: 39648199 PMCID: PMC11626751 DOI: 10.1186/s13046-024-03244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/30/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Liver metastasis poses a significant barrier to effective immunotherapy in patients with colorectal cancer. Cryoablation has emerged as a vital supplementary therapeutic approach for these patients. However, its impact on the tumor microenvironment following the ablation of liver metastases remains unclear. METHODS We acquired multi-omics time-series data at 1 day, 5 days, and 14 days post-cryoablation, based on tumor and peripheral blood samples from clinical patients, cell co-culture models, and a liver metastases mouse model built on the MC38 cell line in C57BL/6 J mice. This dataset included single-cell transcriptomic sequencing, bulk tissue transcriptomic sequencing, 4D-Label-Free proteomics, flow cytometry data, western blot data, and histological immunofluorescence staining of pathological specimens. RESULTS We found that a neutrophil-related inflammatory state persisted for at least 14 days post-cryoablation. During this period, neutrophils underwent phenotypic changes, shifting from the N1 to the N2 type. Cryoablation also caused a significant increase in intracellular Ca2+ concentration in neutrophils, which triggered the formation of PAD4-dependent neutrophil extracellular traps (NETs), further promoting immune evasion. Moreover, animal studies demonstrated that depleting or inhibiting the CXCL2-CXCR2 signaling axis within neutrophils, or degrading NETs, could effectively restore the host's anti-tumor immune response. CONCLUSIONS These findings underscore the critical role of neutrophils and their NETs in immune escape following cryoablation. Targeting the CXCL2-CXCR2-Ca2+-PAD4 axis could enhance the therapeutic response to PD-1 antibodies, providing a potential strategy to improve treatment outcomes for colorectal cancer with liver metastases.
Collapse
Affiliation(s)
- Hongtong Tan
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yiquan Jiang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lujun Shen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Gulijiayina Nuerhashi
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chunyong Wen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ling Gu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yujia Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Han Qi
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fei Cao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tao Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weining Xie
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Guangdong, China
| | - Wuguo Deng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Weijun Fan
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
4
|
Mukherjee D, Satyavolu S, Cioffi S, Thomas A, Li Y, Nayak L. Protocols for circulating neutrophil depletion in neonatal C57Bl/6 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592835. [PMID: 39282419 PMCID: PMC11398316 DOI: 10.1101/2024.05.06.592835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Murine neonatal neutrophil depletion strategies have problems achieving deep neutrophil clearance and accurate residual neutrophil fraction detection. An isotype switch method can achieve profound neutrophil clearance using a combination of anti-Ly6G and anti-rat κ Ig light chain antibodies in adult C57Bl/6 mice, proven by extra- and intracellular Ly6G detection by flow cytometry. We adapted this technique to neonatal mice, testing four neutrophil depletion strategies in the peripheral circulation, bone marrow, and spleen. Four protocols were tested: P3 Ly6G and P1-3 Ly6G (anti-Ly6G on postnatal days (P) 3 and 1-3 respectively), and P3 Dual and P1-3 Dual (anti-Ly6G and anti-rat κ Ig light chain on P3 and P1-3 respectively). Intracellular and extracellular Ly6G presence was detected using flow cytometry. Isotype control antibodies were used as controls. P1-3 Dual protocol achieved significantly better neutrophil depletion than the P1-3 Ly6G or P3 Ly6G protocols (97% vs. 74% and 97% vs. 50%, respectively) in the peripheral circulation. The P3 Dual protocol alone was enough to achieve significantly better neutrophil clearance (93%) than any of the Ly6G alone protocols. The Ly6G alone protocols led to near-total elimination of extracellular Ly6G. However, there was a significant presence of intracellular Ly6G in the CD45+ cell population, evading detection by extracellular Ly6G antibody-based detection methods. P3 protocols perform better than P1-3 protocols for bone marrow and splenic neutrophil clearance. Thus, the P3 Dual protocol might be the most effective and ethical protocol to induce profound neutrophil depletion in neonatal mice, an alternative to daily anti-Ly6G injections. Summary Sentence Dual antibody-based neutrophil depletion effectively induces circulating neutrophil clearance in neonatal mice.
Collapse
|
5
|
Liu Y, Guo F, Han Z, Yin Y, Chen G, Zhang Y, Tang Q, Chen L. Neutrophils inhibit bone formation by directly contacting osteoblasts and suppressing osteogenic differentiation. Bone 2024; 190:117310. [PMID: 39477179 DOI: 10.1016/j.bone.2024.117310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024]
Abstract
Neutrophils have been extensively studied for their critical roles in supporting immune defense mechanisms, initiating bone regeneration, and promoting angiogenesis. Nonetheless, the influence of neutrophils on physiological conditions, particularly in the context of bone development, remains incompletely understood. In this study, we examined the effects of non-inflammatory neutrophils on bone physiology by depleting Ly6G+ neutrophils and inducing neutropenia through myelosuppression. Our results demonstrated a notable increase in bone mass and a decrease in the bone marrow cavity upon depletion of the neutrophils. These effects were attributed to the direct interaction between neutrophils and osteoblasts, independent of reduced secretion of typical inflammatory cytokines or diminished osteoclast differentiation. This observation suggests a non-inflammatory function of neutrophils within the endosteal microenvironment, where they regulate osteogenic differentiation to preserve optimal bone mass, shape healthy three-dimensional bone trabecular structures, and create ample space for hematopoietic niche development.
Collapse
Affiliation(s)
- Yijun Liu
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Fengyuan Guo
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Zhenshuo Han
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Ying Yin
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Guangjin Chen
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yifan Zhang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Qingming Tang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| | - Lili Chen
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| |
Collapse
|
6
|
Whitaker R, Sung S, Tylek T, Risser G, O'Brien E, Chua PE, Li T, Petrie RJ, Han L, Binder-Markey B, Spiller KL. Effects of injury size on local and systemic immune cell dynamics in volumetric muscle loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609702. [PMID: 39253495 PMCID: PMC11383310 DOI: 10.1101/2024.08.26.609702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
We took a systems approach to the analysis of macrophage phenotype in regenerative and fibrotic volumetric muscle loss outcomes in mice together with analysis of systemic inflammation and of other leukocytes in the muscle, spleen, and bone marrow. Macrophage dysfunction in the fibrotic group occurred as early as day 1, persisted to at least day 28, and was associated with increased numbers of leukocytes in the muscle and bone marrow, increased pro-inflammatory marker expression in splenic macrophages, and changes in the levels of pro-inflammatory cytokines in the blood. The most prominent differences were in muscle neutrophils, which were much more abundant in fibrotic outcomes compared to regenerative outcomes at day 1 after injury. However, neutrophil depletion had little to no effect on macrophage phenotype or on muscle repair outcomes. Together, these results suggest that the entire system of immune cell interactions must be considered to improve muscle repair outcomes.
Collapse
Affiliation(s)
- Ricardo Whitaker
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Samuel Sung
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Tina Tylek
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Gregory Risser
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Erin O'Brien
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Phoebe Ellin Chua
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Thomas Li
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Ryan J Petrie
- Department of Biology, College of Arts & Sciences, Drexel University, Philadelphia, PA
| | - Lin Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - Benjamin Binder-Markey
- Department of Physical Therapy & Rehabilitation Sciences, College of Nursing and Health Professions, Drexel University, Philadelphia, PA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| |
Collapse
|
7
|
Zhang Y, Xu J, Miranda-Katz M, Sojati J, Tollefson SJ, Manni ML, Alcorn JF, Sarkar SN, Williams JV. Distinct roles for type I and type III interferons in virulent human metapneumovirus pathogenesis. PLoS Pathog 2024; 20:e1011840. [PMID: 38315735 PMCID: PMC10868789 DOI: 10.1371/journal.ppat.1011840] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/15/2024] [Accepted: 11/17/2023] [Indexed: 02/07/2024] Open
Abstract
Human metapneumovirus (HMPV) is an important cause of acute lower respiratory infection in children and adults worldwide. There are four genetic subgroups of HMPV and both neutralizing antibodies and T cells contribute to protection. However, little is known about mechanisms of pathogenesis and most published work is based on a few extensively passaged, laboratory-adapted strains of HMPV. In this study, we isolated and characterized a panel of low passage HMPV clinical isolates representing all four genetic subgroups. The clinical isolates exhibited lower levels of in vitro replication compared to a lab-adapted strain. We compared disease phenotypes using a well-established mouse model. Several virulent isolates caused severe weight loss, lung pathology, airway dysfunction, and fatal disease in mice, which was confirmed in three inbred mouse strains. Disease severity did not correlate with lung viral titer, as virulent strains exhibited restricted replication in the lower airway. Virulent HMPV isolates were associated with markedly increased proinflammatory cytokine production and neutrophil influx; however, depletion of neutrophils or genetic ablation of inflammasome components did not reverse disease. Virulent clinical isolates induced markedly increased type I and type III interferon (IFN) secretion in vitro and in vivo. STAT1/2-deficient mice lacking both type I and type III IFN signaling showed reduced disease severity and increased lung viral replication. Inhibition of type I IFN signaling using a blocking antibody or genetic ablation of the type I IFN receptor reduced pathology with minimal effect on viral replication. Conversely, blockade of type III IFN signaling with a neutralizing antibody or genetic ablation of the IFN-lambda receptor had no effect on pathogenesis but restored viral replication. Collectively, these results demonstrate distinct roles for type I and type III IFN in HMPV pathogenesis and immunity.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jiuyang Xu
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Tsinghua University School of Medicine, Beijing, China
| | - Margot Miranda-Katz
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jorna Sojati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sharon J. Tollefson
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Michelle L. Manni
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John F. Alcorn
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Saumendra N. Sarkar
- Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pennsylvania, United States of America
| | - John V. Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pennsylvania, United States of America
- Institute for Infection, Inflammation, and Immunity in Children, University of Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
8
|
Chadwick C, De Jesus M, Ginty F, Martinez JS. Pathobiology of Candida auris infection analyzed by multiplexed imaging and single cell analysis. PLoS One 2024; 19:e0293011. [PMID: 38232081 DOI: 10.1371/journal.pone.0293011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 10/03/2023] [Indexed: 01/19/2024] Open
Abstract
Fungal organisms contribute to significant human morbidity and mortality and Candida auris (C. auris) infections are of utmost concern due to multi-drug resistant strains and persistence in critical care and hospital settings. Pathogenesis and pathology of C. auris is still poorly understood and in this study, we demonstrate how the use of multiplex immunofluorescent imaging (MxIF) and single-cell analysis can contribute to a deeper understanding of fungal infections within organs. We used two different neutrophil depletion murine models (treated with either 1A8-an anti-Ly6G antibody, or RB6-8C5-an anti-Ly6G/Ly6C antibody; both 1A8 and RB6-8C5 antibodies have been shown to deplete neutrophils) and compared to wildtype, non-neutropenic mice. Following pathologist assessment, fixed samples underwent MxIF imaging using a C. albicans antibody (shown to be cross-reactive to C. auris) and immune cell biomarkers-CD3 (T cells), CD68 (macrophages), B220 (B cells), CD45 (monocytes), and Ly6G (neutrophils) to quantify organ specific immune niches. MxIF analysis highlighted the heterogenous distribution of C. auris infection within heart, kidney, and brain 7 days post-infection. Size and number of fungal abscesses was greatest in the heart and lowest in brain. Infected mice had an increased count of CD3+, CD68+, B220+, and CD45+ immune cells, concentrated around C. auris abscesses. CD68+ cells were predominant in wildtype (non-neutropenic mice) and CD3+/CD45+ cells were predominant in neutropenic mice, with B cells being the least abundant. These findings suggest a Th2 driven immune response in neutropenic C. auris infection mice models. This study demonstrates the value of MxIF to broaden understanding of C. auris pathobiology, and mechanistic understanding of fungal infections.
Collapse
Affiliation(s)
| | - Magdia De Jesus
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, United States of America
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Fiona Ginty
- GE Research, Niskayuna, New York, United States of America
| | | |
Collapse
|
9
|
Hong X, Luo AC, Doulamis I, Oh N, Im GB, Lin CY, del Nido PJ, Lin RZ, Melero-Martin JM. Photopolymerizable Hydrogel for Enhanced Intramyocardial Vascular Progenitor Cell Delivery and Post-Myocardial Infarction Healing. Adv Healthc Mater 2023; 12:e2301581. [PMID: 37611321 PMCID: PMC10840685 DOI: 10.1002/adhm.202301581] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/08/2023] [Indexed: 08/25/2023]
Abstract
Cell transplantation success for myocardial infarction (MI) treatment is often hindered by low engraftment due to washout effects during myocardial contraction. A clinically viable biomaterial that enhances cell retention can optimize intramyocardial cell delivery. In this study, a therapeutic cell delivery method is developed for MI treatment utilizing a photocrosslinkable gelatin methacryloyl (GelMA) hydrogel. Human vascular progenitor cells, capable of forming functional vasculatures upon transplantation, are combined with an in situ photopolymerization approach and injected into the infarcted zones of mouse hearts. This strategy substantially improves acute cell retention and promotes long-term post-MI cardiac healing, including stabilized cardiac functions, preserved viable myocardium, and reduced cardiac fibrosis. Additionally, engrafted vascular cells polarize recruited bone marrow-derived neutrophils toward a non-inflammatory phenotype via transforming growth factor beta (TGFβ) signaling, fostering a pro-regenerative microenvironment. Neutrophil depletion negates the therapeutic benefits generated by cell delivery in ischemic hearts, highlighting the essential role of non-inflammatory, pro-regenerative neutrophils in cardiac remodeling. In conclusion, this GelMA hydrogel-based intramyocardial vascular cell delivery approach holds promise for enhancing the treatment of acute myocardial infarction.
Collapse
Affiliation(s)
- Xuechong Hong
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Allen Chilun Luo
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ilias Doulamis
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Oh
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Gwang-Bum Im
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Yen Lin
- Department of Lymphoma and Myeloma, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Pedro J. del Nido
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
10
|
He Y, Heng Y, Qin Z, Wei X, Wu Z, Qu J. Intravital microscopy of satellite cell dynamics and their interaction with myeloid cells during skeletal muscle regeneration. SCIENCE ADVANCES 2023; 9:eadi1891. [PMID: 37851799 PMCID: PMC10584350 DOI: 10.1126/sciadv.adi1891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/15/2023] [Indexed: 10/20/2023]
Abstract
Skeletal muscle regeneration requires the highly coordinated cooperation of muscle satellite cells (MuSCs) with other cellular components. Upon injury, myeloid cells populate the wound site, concomitant with MuSC activation. However, detailed analysis of MuSC-myeloid cell interaction is hindered by the lack of suitable live animal imaging technology. Here, we developed a dual-laser multimodal nonlinear optical microscope platform to study the dynamics of MuSCs and their interaction with nonmyogenic cells during muscle regeneration. Using three-dimensional time-lapse imaging on live reporter mice and taking advantages of the autofluorescence of reduced nicotinamide adenine dinucleotide (NADH), we studied the spatiotemporal interaction between nonmyogenic cells and muscle stem/progenitor cells during MuSC activation and proliferation. We discovered that their cell-cell contact was transient in nature. Moreover, MuSCs could activate with notably reduced infiltration of neutrophils and macrophages, and their proliferation, although dependent on macrophages, did not require constant contact with them. These findings provide a fresh perspective on myeloid cells' role during muscle regeneration.
Collapse
Affiliation(s)
- Yingzhu He
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| | - Youshan Heng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| | - Zhongya Qin
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| | - Xiuqing Wei
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| | - Zhenguo Wu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| | - Jianan Qu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
11
|
Complement, but Not Platelets, Plays a Pivotal Role in the Outcome of Mucormycosis In Vivo. J Fungi (Basel) 2023; 9:jof9020162. [PMID: 36836277 PMCID: PMC9965864 DOI: 10.3390/jof9020162] [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: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Mucormycetes, a heterogeneous group of fungi, induce a life-threatening disease called mucormycosis. Immune deficiencies represent a major risk factor; hence, we wanted to illuminate the role of complement and platelets in the defense against mucormycetes. METHODS Rhizopus arrhizus (Ra), Rhizopus microsporus (Rm), Lichtheimia ramosa (Lr), Lichtheimia corymbifera (Lc), Rhizomucor pusillus (Rmp), and Mucor circinelloides (Mc) spores were opsonized with human and mouse serum, and C1q, C3c, and terminal complement complex (C5b-9) deposition was measured. Additionally, thrombocytopenic, C3-deficient, or C6-deficient mice were intravenously infected with selected isolates. Survival and immunological parameters were monitored, and fungal burden was determined and compared to that of immunocompetent and neutropenic mice. RESULTS In vitro experiments showed significant differences in complement deposition between mucormycetes. Mc isolates bound up to threefold more human C5b-9 than other mucormycetes. Lr, Lc, and Mc bound high levels of murine C3c, whereas human C3c deposition was reduced on Mc compared to Lr and Lc. Murine C3c deposition negatively correlated with virulence. Complement deficiencies and neutropenia, but not thrombocytopenia, were shown to be a risk factor for a lethal outcome. CONCLUSION Complement deposition varies between mucormycetes. Additionally, we demonstrated that complement and neutrophilic granulocytes, but not platelets, play an important role in a murine model of disseminated mucormycosis.
Collapse
|
12
|
Sieow JL, Penny HL, Gun SY, Tan LQ, Duan K, Yeong JPS, Pang A, Lim D, Toh HC, Lim TKH, Engleman E, Rotzschke O, Ng LG, Chen J, Tan SM, Wong SC. Conditional Knockout of Hypoxia-Inducible Factor 1-Alpha in Tumor-Infiltrating Neutrophils Protects against Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2023; 24:ijms24010753. [PMID: 36614196 PMCID: PMC9821271 DOI: 10.3390/ijms24010753] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 01/04/2023] Open
Abstract
Large numbers of neutrophils infiltrate tumors and comprise a notable component of the inflammatory tumor microenvironment. While it is established that tumor cells exhibit the Warburg effect for energy production, the contribution of the neutrophil metabolic state to tumorigenesis is unknown. Here, we investigated whether neutrophil infiltration and metabolic status promotes tumor progression in an orthotopic mouse model of pancreatic ductal adenocarcinoma (PDAC). We observed a large increase in the proportion of neutrophils in the blood and tumor upon orthotopic transplantation. Intriguingly, these tumor-infiltrating neutrophils up-regulated glycolytic factors and hypoxia-inducible factor 1-alpha (HIF-1α) expression compared to neutrophils from the bone marrow and blood of the same mouse. This enhanced glycolytic signature was also observed in human PDAC tissue samples. Strikingly, neutrophil-specific deletion of HIF-1α (HIF-1αΔNφ) significantly reduced tumor burden and improved overall survival in orthotopic transplanted mice, by converting the pro-tumorigenic neutrophil phenotype to an anti-tumorigenic phenotype. This outcome was associated with elevated reactive oxygen species production and activated natural killer cells and CD8+ cytotoxic T cells compared to littermate control mice. These data suggest a role for HIF-1α in neutrophil metabolism, which could be exploited as a target for metabolic modulation in cancer.
Collapse
Affiliation(s)
- Je Lin Sieow
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Hweixian Leong Penny
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Sin Yee Gun
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Ling Qiao Tan
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Kaibo Duan
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Joe Poh Sheng Yeong
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Angela Pang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| | - Diana Lim
- Department of Pathology, National University Health System, Singapore 119074, Singapore
| | - Han Chong Toh
- Department of Oncology, National Cancer Centre, Singapore 169610, Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Edgar Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Olaf Rotzschke
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Suet Mien Tan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Siew Cheng Wong
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
- Correspondence: ; Tel.: +65-64070030
| |
Collapse
|
13
|
Wu X, You D, Cui J, Yang L, Lin L, Chen Y, Xu C, Lian G, Wan J. Reduced Neutrophil Extracellular Trap Formation During Ischemia Reperfusion Injury in C3 KO Mice: C3 Requirement for NETs Release. Front Immunol 2022; 13:781273. [PMID: 35250972 PMCID: PMC8889019 DOI: 10.3389/fimmu.2022.781273] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/20/2022] [Indexed: 12/27/2022] Open
Abstract
Complement C3 plays a prominent role in inflammatory processes, and its increase exacerbates ischemia reperfusion injury (IRI)-induced acute kidney injury (AKI). Infiltrated neutrophils can be stimulated to form neutrophil extracellular traps (NETs), leading to renal injury. However, the relationship between the increase of C3 and the release of NETs in AKI was not clear. Here we found that IRI in the mouse kidney leads to increased neutrophils infiltration and NET formation. Furthermore, neutrophils depletion by anti-Ly6G IgG (1A8) did not reduce C3 activation but reduced kidney injury and inflammation, indicating a link between neutrophils infiltration and renal tissue damage. Pretreatment with 1A8 suppressed ischemia-induced NET formation, proving that extracellular traps (ETs) in renal tissue were mainly derived from neutrophils. Renal ischemia injury also leads to increased expression of C3. Moreover, C3 KO mice (C3 KO) with IRI exhibited attenuated kidney damage and decreased neutrophils and NETs. In vitro, C3a primed neutrophils to form NETs, reflected by amorphous extracellular DNA structures that colocalized with CitH3 and MPO. These data reveal that C3 deficiency can ameliorate AKI by reducing the infiltration of neutrophils and the formation of NETs. Targeting C3 activation may be a new therapeutic strategy for alleviating the necroinflammation of NETs in AKI.
Collapse
Affiliation(s)
- Xiaoting Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Danyu You
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jiong Cui
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Liyan Yang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Liyu Lin
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yi Chen
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Changsheng Xu
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guili Lian
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Gunaseelan S, Ariffin MZ, Khanna S, Ooi MH, Perera D, Chu JJH, Chua JJE. Pharmacological perturbation of CXCL1 signaling alleviates neuropathogenesis in a model of HEVA71 infection. Nat Commun 2022; 13:890. [PMID: 35173169 PMCID: PMC8850555 DOI: 10.1038/s41467-022-28533-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 01/27/2022] [Indexed: 12/14/2022] Open
Abstract
Hand, foot and mouth disease (HFMD) caused by Human Enterovirus A71 (HEVA71) infection is typically a benign infection. However, in minority of cases, children can develop severe neuropathology that culminate in fatality. Approximately 36.9% of HEVA71-related hospitalizations develop neurological complications, of which 10.5% are fatal. Yet, the mechanism by which HEVA71 induces these neurological deficits remain unclear. Here, we show that HEVA71-infected astrocytes release CXCL1 which supports viral replication in neurons by activating the CXCR2 receptor-associated ERK1/2 signaling pathway. Elevated CXCL1 levels correlates with disease severity in a HEVA71-infected mice model. In humans infected with HEVA71, high CXCL1 levels are only present in patients presenting neurological complications. CXCL1 release is specifically triggered by VP4 synthesis in HEVA71-infected astrocytes, which then acts via its receptor CXCR2 to enhance viral replication in neurons. Perturbing CXCL1 signaling or VP4 myristylation strongly attenuates viral replication. Treatment with AZD5069, a CXCL1-specific competitor, improves survival and lessens disease severity in infected animals. Collectively, these results highlight the CXCL1-CXCR2 signaling pathway as a potential target against HFMD neuropathogenesis.
Collapse
Affiliation(s)
- Saravanan Gunaseelan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, National University of Singapore, Singapore, 117597, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore
| | - Mohammed Zacky Ariffin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sanjay Khanna
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore
| | - Mong How Ooi
- Department of Paediatrics, Sarawak General Hospital, Kuching, Sarawak, Malaysia
- Institute of Health and Community Medicine, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - David Perera
- Institute of Health and Community Medicine, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Infectious Disease Translational Research Programme, National University of Singapore, Singapore, 117597, Singapore.
| | - John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- LSI Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
15
|
Roy R, Zayas J, Singh SK, Delgado K, Wood SJ, Mohamed MF, Frausto DM, Estupinian R, Giurini EF, Kuzel TM, Zloza A, Reiser J, Shafikhani SH. Overriding impaired FPR chemotaxis signaling in diabetic neutrophil stimulates infection control in murine diabetic wound. eLife 2022; 11:72071. [PMID: 35112667 PMCID: PMC8846594 DOI: 10.7554/elife.72071] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 02/01/2022] [Indexed: 11/19/2022] Open
Abstract
Infection is a major co-morbidity that contributes to impaired healing in diabetic wounds. Although impairments in diabetic neutrophils have been blamed for this co-morbidity, what causes these impairments and whether they can be overcome, remain largely unclear. Diabetic neutrophils, isolated from diabetic individuals, exhibit chemotaxis impairment but this peculiar functional impairment has been largely ignored because it appears to contradict the clinical findings which blame excessive neutrophil influx as a major impediment to healing in chronic diabetic ulcers. Here, we report that exposure to glucose in diabetic range results in impaired chemotaxis signaling through the formyl peptide receptor (FPR) in neutrophils, culminating in reduced chemotaxis and delayed neutrophil trafficking in the wound of Leprdb (db/db) type two diabetic mice, rendering diabetic wound vulnerable to infection. We further show that at least some auxiliary receptors remain functional under diabetic conditions and their engagement by the pro-inflammatory cytokine CCL3, overrides the requirement for FPR signaling and substantially improves infection control by jumpstarting the neutrophil trafficking toward infection, and stimulates healing in diabetic wound. We posit that CCL3 may have therapeutic potential for the treatment of diabetic foot ulcers if it is applied topically after the surgical debridement process which is intended to reset chronic ulcers into acute fresh wounds.
Collapse
Affiliation(s)
- Ruchi Roy
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Janet Zayas
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Sunil K Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, United States
| | - Kaylee Delgado
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Stephen J Wood
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, United States
| | - Mohamed F Mohamed
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Dulce M Frausto
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Ricardo Estupinian
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Eileena F Giurini
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Timothy M Kuzel
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Andrew Zloza
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Sasha H Shafikhani
- Department of Medicine, Rush University Medical Center, Chicago, United States
| |
Collapse
|
16
|
Chao T, Lu L, Zhang L, Huang R, Liu Z, Zhou B, Kong E, Zhang Z, Lawrence T, Liang Y. An inducible model for specific neutrophil depletion by diphtheria toxin in mice. SCIENCE CHINA. LIFE SCIENCES 2021; 64:1227-1235. [PMID: 33420927 DOI: 10.1007/s11427-020-1839-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/15/2020] [Indexed: 06/12/2023]
Abstract
Neutrophils are crucial for immunity and play important roles in inflammatory diseases; however, mouse models selectively deficient in neutrophils are limited, and neutrophil-specific diphtheria toxin (DT)-based depletion system has not yet been established. In this study, we generated a novel knock-in mouse model expressing diphtheria toxin receptor (DTR) under control of the endogenous Ly6G promoter. We showed that DTR expression was restricted to Ly6G+ neutrophils and complete depletion of neutrophils could be achieved by DT treatment at 24-48 h intervals. We characterized the effects of specific neutrophil depletion in mice at steady-state, with acute inflammation and during tumor growth. Our study presents a valuable new tool to study the roles of neutrophils in the immune system and during tumor progression.
Collapse
Affiliation(s)
- Tianzhu Chao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liaoxun Lu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Lichen Zhang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Rong Huang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhuangzhuang Liu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Binhui Zhou
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Eryan Kong
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhongjian Zhang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Toby Lawrence
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, SE1 1UL, UK.
| | - Yinming Liang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China.
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
17
|
Maternal Neutrophil Depletion Fails to Avert Systemic Lipopolysaccharide-Induced Early Pregnancy Defects in Mice. Int J Mol Sci 2021; 22:ijms22157932. [PMID: 34360700 PMCID: PMC8347248 DOI: 10.3390/ijms22157932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 11/17/2022] Open
Abstract
Maternal infection-induced early pregnancy complications arise from perturbation of the immune environment at the uterine early blastocyst implantation site (EBIS), yet the underlying mechanisms remain unclear. Here, we demonstrated in a mouse model that the progression of normal pregnancy from days 4 to 6 induced steady migration of leukocytes away from the uterine decidual stromal zone (DSZ) that surrounds the implanted blastocyst. Uterine macrophages were found to be CD206+ M2-polarized. While monocytes were nearly absent in the DSZ, DSZ cells were found to express monocyte marker protein Ly6C. Systemic endotoxic lipopolysaccharide (LPS) exposure on day 5 of pregnancy led to: (1) rapid (at 2 h) induction of neutrophil chemoattractants that promoted huge neutrophil infiltrations at the EBISs by 24 h; (2) rapid (at 2 h) elevation of mRNA levels of MyD88, but not Trif, modulated cytokines at the EBISs; and (3) dose-dependent EBIS defects by day 7 of pregnancy. Yet, elimination of maternal neutrophils using anti-Ly6G antibody prior to LPS exposure failed to avert LPS-induced EBIS defects allowing us to suggest that activation of Tlr4-MyD88 dependent inflammatory pathway is involved in LPS-induced defects at EBISs. Thus, blocking the activation of the Tlr4-MyD88 signaling pathway may be an interesting approach to prevent infection-induced pathology at EBISs.
Collapse
|
18
|
Uhl B, A Mittmann L, Dominik J, Hennel R, Smiljanov B, Haring F, B Schaubächer J, Braun C, Padovan L, Pick R, Canis M, Schulz C, Mack M, Gutjahr E, Sinn P, Heil J, Steiger K, Kanse SM, Weichert W, Sperandio M, Lauber K, Krombach F, Reichel CA. uPA-PAI-1 heteromerization promotes breast cancer progression by attracting tumorigenic neutrophils. EMBO Mol Med 2021; 13:e13110. [PMID: 33998175 PMCID: PMC8185543 DOI: 10.15252/emmm.202013110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 04/28/2021] [Accepted: 03/25/2021] [Indexed: 01/13/2023] Open
Abstract
High intratumoral levels of urokinase-type plasminogen activator (uPA)-plasminogen activator inhibitor-1 (PAI-1) heteromers predict impaired survival and treatment response in early breast cancer. The pathogenetic role of this protein complex remains obscure. Here, we demonstrate that heteromerization of uPA and PAI-1 multiplies the potential of the single proteins to attract pro-tumorigenic neutrophils. To this end, tumor-released uPA-PAI-1 utilizes very low-density lipoprotein receptor and mitogen-activated protein kinases to initiate a pro-inflammatory program in perivascular macrophages. This enforces neutrophil trafficking to cancerous lesions and skews these immune cells toward a pro-tumorigenic phenotype, thus supporting tumor growth and metastasis. Blockade of uPA-PAI-1 heteromerization by a novel small-molecule inhibitor interfered with these events and effectively prevented tumor progression. Our findings identify a therapeutically targetable, hitherto unknown interplay between hemostasis and innate immunity that drives breast cancer progression. As a personalized immunotherapeutic strategy, blockade of uPA-PAI-1 heteromerization might be particularly beneficial for patients with highly aggressive uPA-PAI-1high tumors.
Collapse
Affiliation(s)
- Bernd Uhl
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Laura A Mittmann
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Julian Dominik
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Roman Hennel
- Department of Radiation OncologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Bojan Smiljanov
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Florian Haring
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Johanna B Schaubächer
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Constanze Braun
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Lena Padovan
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Robert Pick
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Martin Canis
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christian Schulz
- Department of CardiologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Matthias Mack
- Department of Internal MedicineUniversity of RegensburgRegensburgGermany
| | - Ewgenija Gutjahr
- Institute for PathologyUniversity of HeidelbergHeidelbergGermany
| | - Peter Sinn
- Institute for PathologyUniversity of HeidelbergHeidelbergGermany
| | - Jörg Heil
- Department of Gynecology and ObstetricsUniversity of HeidelbergHeidelbergGermany
| | - Katja Steiger
- Department of PathologyTechnical University of MunichMunichGermany
| | - Sandip M Kanse
- Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Wilko Weichert
- Department of PathologyTechnical University of MunichMunichGermany
- German Cancer Consortium (DKTK), partner site MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and PathophysiologyLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Kirsten Lauber
- Department of Radiation OncologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Fritz Krombach
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christoph A Reichel
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| |
Collapse
|
19
|
Nielsen TB, Yan J, Luna BM, Talyansky Y, Slarve M, Bonomo RA, Spellberg B. Monoclonal antibody requires immunomodulation for efficacy against Acinetobacter baumannii infection. J Infect Dis 2021; 224:2133-2147. [PMID: 34036366 DOI: 10.1093/infdis/jiab265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Monoclonal antibodies (MAbs) are gaining significant momentum as novel therapeutics for infections caused by antibiotic-resistant bacteria. We evaluated the mechanism by which anti-bacterial MAb therapy protects against Acinetobacter baumannii infections. Anti-capsular MAb enhanced macrophage opsonophagocytosis and rescued mice from lethal infections by harnessing complement, macrophages, and neutrophils, yet the degree of bacterial burden did not correlate with survival. Furthermore, MAb therapy reduced pro-inflammatory (IL-1β, IL-6, TNFα) and anti-inflammatory (IL-10) cytokines, which correlated inversely with survival. Although disrupting IL-10 abrogated the survival advantage conferred by the MAb, IL-10-knockout mice treated with MAb could still survive if TNFα production was suppressed directly (via anti-TNFα neutralizing antibody) or indirectly (via macrophage depletion). Thus, even for a MAb that enhances microbial clearance via opsonophagocytosis, clinical efficacy required modulation of pro- and anti-inflammatory cytokines. These findings may inform future MAb development targeting bacteria that trigger the sepsis cascade.
Collapse
Affiliation(s)
- Travis B Nielsen
- Stritch School of Medicine at Loyola University Chicago, Maywood, IL, 60153, United States.,Department of Medicine and Department of Molecular Microbiology & Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, CA, 90033, United States
| | - Jun Yan
- Department of Medicine and Department of Molecular Microbiology & Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, CA, 90033, United States
| | - Brian M Luna
- Department of Medicine and Department of Molecular Microbiology & Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, CA, 90033, United States
| | - Yuli Talyansky
- Department of Medicine and Department of Molecular Microbiology & Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, CA, 90033, United States
| | - Matthew Slarve
- Department of Medicine and Department of Molecular Microbiology & Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, CA, 90033, United States
| | - Robert A Bonomo
- Department of Medicine, Pharmacology, and Molecular Biology and Microbiology, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Case Western Reserve University, Cleveland, OH, 44120, United States
| | - Brad Spellberg
- Department of Medicine and Department of Molecular Microbiology & Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, CA, 90033, United States
| |
Collapse
|
20
|
Tan X, Petri B, DeVinney R, Jenne CN, Chaconas G. The Lyme disease spirochete can hijack the host immune system for extravasation from the microvasculature. Mol Microbiol 2021; 116:498-515. [PMID: 33891779 DOI: 10.1111/mmi.14728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/30/2022]
Abstract
Lyme disease is the most common tick-transmitted disease in the northern hemisphere and is caused by the spirochete Borrelia burgdorferi and related Borrelia species. The constellation of symptoms attributable to this malady results from vascular dissemination of B. burgdorferi throughout the body to invade various tissue types. However, little is known about the mechanism by which the spirochetes can breach the blood vessel wall to reach distant tissues. We have studied this process by direct observation of spirochetes in the microvasculature of living mice using multi-laser spinning-disk intravital microscopy. Our results show that in our experimental system, instead of phagocytizing B. burgdorferi, host neutrophils are involved in the production of specific cytokines that activate the endothelium and potentiate B. burgdorferi escape into the surrounding tissue. Spirochete escape is not induced by paracellular permeability and appears to occur via a transcellular pathway. Neutrophil repurposing to promote bacterial extravasation represents a new and innovative pathogenic strategy.
Collapse
Affiliation(s)
- Xi Tan
- Department of Biochemistry & Molecular Biology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Björn Petri
- Department of Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Rebekah DeVinney
- Department of Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Craig N Jenne
- Department of Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - George Chaconas
- Department of Biochemistry & Molecular Biology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.,Department of Microbiology, Immunology & Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
21
|
Sugita J, Fujiu K, Nakayama Y, Matsubara T, Matsuda J, Oshima T, Liu Y, Maru Y, Hasumi E, Kojima T, Seno H, Asano K, Ishijima A, Tomii N, Yamazaki M, Kudo F, Sakuma I, Nagai R, Manabe I, Komuro I. Cardiac macrophages prevent sudden death during heart stress. Nat Commun 2021; 12:1910. [PMID: 33771995 PMCID: PMC7997915 DOI: 10.1038/s41467-021-22178-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/27/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiac arrhythmias are a primary contributor to sudden cardiac death, a major unmet medical need. Because right ventricular (RV) dysfunction increases the risk for sudden cardiac death, we examined responses to RV stress in mice. Among immune cells accumulated in the RV after pressure overload-induced by pulmonary artery banding, interfering with macrophages caused sudden death from severe arrhythmias. We show that cardiac macrophages crucially maintain cardiac impulse conduction by facilitating myocardial intercellular communication through gap junctions. Amphiregulin (AREG) produced by cardiac macrophages is a key mediator that controls connexin 43 phosphorylation and translocation in cardiomyocytes. Deletion of Areg from macrophages led to disorganization of gap junctions and, in turn, lethal arrhythmias during acute stresses, including RV pressure overload and β-adrenergic receptor stimulation. These results suggest that AREG from cardiac resident macrophages is a critical regulator of cardiac impulse conduction and may be a useful therapeutic target for the prevention of sudden death.
Collapse
MESH Headings
- Amphiregulin/metabolism
- Amphiregulin/physiology
- Animals
- Animals, Newborn
- Arrhythmias, Cardiac/complications
- Cells, Cultured
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/prevention & control
- Female
- Gap Junctions/physiology
- HeLa Cells
- Humans
- Macrophages/metabolism
- Macrophages/physiology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/cytology
- Myocardium/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Receptors, Adrenergic, beta/metabolism
- Mice
Collapse
Affiliation(s)
- Junichi Sugita
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Katsuhito Fujiu
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Department of Advanced Cardiology, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama, 332-0012, Japan.
| | - Yukiteru Nakayama
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takumi Matsubara
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Jun Matsuda
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tsukasa Oshima
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yuxiang Liu
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yujin Maru
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Eriko Hasumi
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Toshiya Kojima
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroshi Seno
- Medical Device Development and Regulation Research Center, Department of Bioengineering/Department of Precision Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Keisuke Asano
- Medical Device Development and Regulation Research Center, Department of Bioengineering/Department of Precision Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ayumu Ishijima
- Medical Device Development and Regulation Research Center, Department of Bioengineering/Department of Precision Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Naoki Tomii
- Medical Device Development and Regulation Research Center, Department of Bioengineering/Department of Precision Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Masatoshi Yamazaki
- Medical Device Development and Regulation Research Center, Department of Bioengineering/Department of Precision Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Fujimi Kudo
- Department of Disease Biology and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Ichiro Sakuma
- Medical Device Development and Regulation Research Center, Department of Bioengineering/Department of Precision Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ryozo Nagai
- Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi-ken, Tochigi, 329-0498, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.
| | - Issei Komuro
- Department of Cardiovascular Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
22
|
Seo DH, Che X, Kim S, Kim DH, Ma HW, Kim JH, Kim TI, Kim WH, Kim SW, Cheon JH. Triggering Receptor Expressed on Myeloid Cells-1 Agonist Regulates Intestinal Inflammation via Cd177 + Neutrophils. Front Immunol 2021; 12:650864. [PMID: 33767714 PMCID: PMC7985452 DOI: 10.3389/fimmu.2021.650864] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
Triggering receptor expressed on myeloid cell-1 (TREM-1) signaling is expressed on neutrophils and monocytes that is necessary for the successful antimicrobial response and resolution of inflammation in the gut. In this study, we determined the effect of an anti-TREM-1 agonistic antibody (α-TREM-1) on colitis and identify its underlying mechanism of action. Administration of α-TREM-1 alleviated colitis in mice and resolved dysbiosis, which required TLR4/Myd88 signaling. α-TREM-1 increased the production of neutrophil extracellular traps and interleukin-22 by CD177+ neutrophils, which led to pathogen clearance and protection of the intestinal barrier. TREM-1 activation using an α-TREM-1 antibody protects against colitis by rebalancing the microbiota and protecting the epithelium against the immune response as well as modulates the function of neutrophils and macrophages. These results highlight the importance of the TREM-1 pathway in intestinal homeostasis and suggest that α-TREM-1 treatment may be an effective therapeutic strategy for inflammatory bowel disease.
Collapse
Affiliation(s)
- Dong Hyuk Seo
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Xiumei Che
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Soochan Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Da Hye Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Hyeon Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Il Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Won Ho Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
23
|
Erdener ŞE, Tang J, Kılıç K, Postnov D, Giblin JT, Kura S, Chen ICA, Vayisoğlu T, Sakadžić S, Schaffer CB, Boas DA. Dynamic capillary stalls in reperfused ischemic penumbra contribute to injury: A hyperacute role for neutrophils in persistent traffic jams. J Cereb Blood Flow Metab 2021; 41:236-252. [PMID: 32237951 PMCID: PMC8370003 DOI: 10.1177/0271678x20914179] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Ever since the introduction of thrombolysis and the subsequent expansion of endovascular treatments for acute ischemic stroke, it remains to be identified why the actual outcomes are less favorable despite recanalization. Here, by high spatio-temporal resolution imaging of capillary circulation in mice, we introduce the pathological phenomenon of dynamic flow stalls in cerebral capillaries, occurring persistently in salvageable penumbra after reperfusion. These stalls, which are different from permanent cellular plugs of no-reflow, were temporarily and repetitively occurring in the capillary network, impairing the overall circulation like small focal traffic jams. In vivo microscopy in the ischemic penumbra revealed leukocytes traveling slowly through capillary lumen or getting stuck, while red blood cell flow was being disturbed in the neighboring segments under reperfused conditions. Stall dynamics could be modulated, by injection of an anti-Ly6G antibody specifically targeting neutrophils. Decreased number and duration of stalls were associated with improvement in penumbral blood flow within 2-24 h after reperfusion along with increased capillary oxygenation, decreased cellular damage and improved functional outcome. Thereby, dynamic microcirculatory stall phenomenon can be a contributing factor to ongoing penumbral injury and is a potential hyperacute mechanism adding on previous observations of detrimental effects of activated neutrophils in ischemic stroke.
Collapse
Affiliation(s)
- Şefik E Erdener
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA.,Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jianbo Tang
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA.,Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Kıvılcım Kılıç
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dmitry Postnov
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA.,Institute of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - John T Giblin
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sreekanth Kura
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - I-Chun A Chen
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Tuğberk Vayisoğlu
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sava Sakadžić
- Optics Division, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - David A Boas
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
24
|
Shi Y, Tomczak K, Li J, Ochieng JK, Lee Y, Haymaker C. Next-Generation Immunotherapies to Improve Anticancer Immunity. Front Pharmacol 2021; 11:566401. [PMID: 33505304 PMCID: PMC7831045 DOI: 10.3389/fphar.2020.566401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Checkpoint inhibitors are widely used immunotherapies for advanced cancer. Nonetheless, checkpoint inhibitors have a relatively low response rate, work in a limited range of cancers, and have some unignorable side effects. Checkpoint inhibitors aim to reinvigorate exhausted or suppressed T cells in the tumor microenvironment (TME). However, the TME contains various other immune cell subsets that interact to determine the fate of cytotoxic T cells. Activation of cytotoxic T cells is initiated by antigen cross-presentation of dendritic cells. Dendritic cells could also release chemokines and cytokines to recruit and foster T cells. B cells, another type of antigen-presenting cell, also foster T cells and can produce tumor-specific antibodies. Neutrophils, a granulocyte cell subset in the TME, impede the proliferation and activation of T cells. The TME also consists of cytotoxic innate natural killer cells, which kill tumor cells efficiently. Natural killer cells can eradicate major histocompatibility complex I-negative tumor cells, which escape cytotoxic T cell–mediated destruction. A thorough understanding of the immune mechanism of the TME, as reviewed here, will lead to further development of more powerful therapeutic strategies. We have also reviewed the clinical outcomes of patients treated with drugs targeting these immune cells to identify strategies for improvement and possible immunotherapy combinations.
Collapse
Affiliation(s)
- Yaoyao Shi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katarzyna Tomczak
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - June Li
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joshua K Ochieng
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Younghee Lee
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
25
|
Lovewell RR, Baer CE, Mishra BB, Smith CM, Sassetti CM. Granulocytes act as a niche for Mycobacterium tuberculosis growth. Mucosal Immunol 2021; 14:229-241. [PMID: 32483198 PMCID: PMC7704924 DOI: 10.1038/s41385-020-0300-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/10/2020] [Accepted: 04/23/2020] [Indexed: 02/04/2023]
Abstract
Granulocyte recruitment to the pulmonary compartment is a hallmark of progressive tuberculosis (TB). This process is well-documented to promote immunopathology, but can also enhance the replication of the pathogen. Both the specific granulocytes responsible for increasing mycobacterial burden and the underlying mechanisms remain obscure. We report that the known immunomodulatory effects of these cells, such as suppression of protective T-cell responses, play a limited role in altering host control of mycobacterial replication in susceptible mice. Instead, we find that the adaptive immune response preferentially restricts the burden of bacteria within monocytes and macrophages compared to granulocytes. Specifically, mycobacteria within inflammatory lesions are preferentially found within long-lived granulocytes that express intermediate levels of the Ly6G marker and low levels of antimicrobial genes. These cells progressively accumulate in the lung and correlate with bacterial load and disease severity, and the ablation of Ly6G-expressing cells lowers mycobacterial burden. These observations suggest a model in which dysregulated granulocytic influx promotes disease by creating a permissive intracellular niche for mycobacterial growth and persistence.
Collapse
Affiliation(s)
- Rustin R Lovewell
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Christina E Baer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Bibhuti B Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Clare M Smith
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
26
|
Joekel DE, Nur S, Monné Rodriguez J, Kronenberg PA, Kipar A, LeibundGut-Landmann S, Deplazes P. Agranulocytosis leads to intestinal Echinococcus multilocularis oncosphere invasion and hepatic metacestode development in naturally resistant Wistar rats. Parasitology 2021; 148:53-62. [PMID: 33087186 PMCID: PMC11010136 DOI: 10.1017/s0031182020002012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022]
Abstract
Susceptibility to Echinococcus multilocularis infection considerably varies among intermediate (mostly rodents) and dead-end host species (e.g. humans and pig), in particular regarding intestinal oncosphere invasion and subsequent hepatic metacestode development. Wistar rats are highly resistant to infection and subsequent diseases upon oral inoculation with E. multilocularis eggs, however, after immunosuppressive treatment with dexamethasone, rats become susceptible. To address the role of the cellular innate immunity, Wistar rats were individually or combined depleted of natural killer (NK) cells, macrophages (MΦ) and granulocytes (polymorphonuclear cells, PMN) prior to E. multilocularis egg inoculation. Although NK cell and MΦ depletion did not alter the resistance status of rats, the majority of PMN-depleted animals developed liver metacestodes within 10 weeks, indicating that PMN are key players in preventing oncosphere migration and/or development in Wistar rats. In vitro studies indicated that resistance is not caused by neutrophil reactive oxygen species or NETosis. Also, light microscopical examinations of the small intestine showed that oral inoculation of E. multilocularis eggs does not elicit a mucosal neutrophil response, suggesting that the interaction of oncospheres and neutrophils may occur after the former have entered the peripheral blood. We suggest to consider granulocytes as mediators of resistance in more resistant species, such as humans.
Collapse
Affiliation(s)
- Deborah E. Joekel
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
| | - Selim Nur
- Section of Immunology, Institute of Virology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057Zurich, Switzerland
| | - Josep Monné Rodriguez
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, 8057Zurich, Switzerland
| | - Philipp A. Kronenberg
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012Bern, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, 8057Zurich, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Institute of Virology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057Zurich, Switzerland
| | - Peter Deplazes
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
| |
Collapse
|
27
|
Osna NA, Bhatia R, Thompson C, Batra SK, Kumar S, Cho Y, Szabo G, Molina PE, Weinman SA, Ganesan M, Kharbanda KK. Role of non-Genetic Risk Factors in Exacerbating Alcohol-related organ damage. Alcohol 2020; 87:63-72. [PMID: 32497558 PMCID: PMC7483997 DOI: 10.1016/j.alcohol.2020.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 02/08/2023]
Abstract
This review provides a summary of the symposium titled "Role of Non-Genetic Risk Factors in Exacerbating Alcohol-Related Organ Damage", which was held at the 42nd Annual Meeting of the Research Society on Alcoholism. The goals of the symposium were to provide newer insights into the role of non-genetic factors, including specific external factors, notably infectious agents or lifestyle factors, that synergistically act to exacerbate alcohol pathogenicity to generate more dramatic downstream biological defects. This summary of the symposium will benefit junior/senior basic scientists and clinicians currently investigating/treating alcohol-induced organ pathology, as well as undergraduate, graduate, and post-graduate students and fellows.
Collapse
Affiliation(s)
- Natalia A Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Rakesh Bhatia
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Christopher Thompson
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Surinder K Batra
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sushil Kumar
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yeonhee Cho
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Patricia E Molina
- Department of Physiology, LSUHSC-New Orleans, New Orleans, LA, United States
| | - Steven A Weinman
- Department of Internal Medicine and the Liver Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States; Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
28
|
Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer 2020; 20:485-503. [PMID: 32694624 DOI: 10.1038/s41568-020-0281-y] [Citation(s) in RCA: 687] [Impact Index Per Article: 137.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.
Collapse
Affiliation(s)
- Sebastien Jaillon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
| | - Andrea Ponzetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Diletta Di Mitri
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Angela Santoni
- Dipartimento di Medicina Molecolare Istituto Pasteur-Fondazione Cenci Bolognetti, Università di Roma 'La Sapienza', Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
- The William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
29
|
Mert T, Sahin E, Yaman S, Sahin M. Effects of immune cell-targeted treatments result from the suppression of neuronal oxidative stress and inflammation in experimental diabetic rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1293-1302. [PMID: 32361779 DOI: 10.1007/s00210-020-01871-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/08/2020] [Indexed: 01/17/2023]
Abstract
In this study, we hypothesized that reduction of immune cell activation as well as their oxidant or inflammatory mediators with minocycline (MCN), liposome-encapsulated clodronate (LEC), or anti-Ly6G treatments can be neuroprotective approaches in diabetic neuropathy. MCN (40 mg/kg) for reduction of microglial activation, LEC (25 mg/kg) for of macrophage inhibition, or anti-Ly6G (150 μg/kg) for neutrophil suppression injected to streptozotocin (STZ)-induced diabetic rats twice, 3 days, and 1 week (half dose) after STZ. Animal mass and blood glucose levels were measured; thermal and mechanical sensitivities were tested for in pain sensations. The levels of chemokine C-X-C motif ligand 1 (CXCL1), CXCL8, and C-C motif ligand 2 (CCL2), CCL3, and total oxidant status (TOS) and total antioxidant status (TAS) were measured in the spinal cord and sciatic nerve tissues of rats. LEC significantly reduced the glucose level of diabetic rats compared with drug control. However, MCN or anti-LY6G did not change the glucose level. While diabetic rats showed a marked decrease in both thermal and mechanical sensations, all treatments alleviated these abnormal sensations. The levels of chemokines and oxidative stress parameters increased in diabetic rats. All drug treatments significantly decreased the CCL2, CXCL1, and CXCL8 levels of spinal cord tissues and ameliorated the neuronal oxidative stress compared with control treatments. Present findings suggest that the neuroprotective actions of MCN, LEC, or anti-Ly6G treatments may be due to the modulation of neuronal oxidative stress and/or inflammatory mediators of immune cells in diabetic rats with neuropathy.
Collapse
Affiliation(s)
- Tufan Mert
- Department of Biophysics, Faculty of Medicine, Bolu Abant Izzet Baysal University, 14030, Bolu, Turkey.
| | - Emel Sahin
- Department of Medical Biology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Selma Yaman
- Department of Biophysics, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Mehmet Sahin
- Department of Medical Biology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
30
|
Mert T, Metin TO, Sahin M, Yaman S. Antiinflammatory properties of antiLy6G antibody disappear during magnetic field exposure in rats with carrageenan induced acute paw inflammation. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:2107-2115. [PMID: 32592030 DOI: 10.1007/s00210-020-01925-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/17/2020] [Indexed: 12/01/2022]
Abstract
Antiinflammatory properties of pulsed magnetic field (PMF) treatments or administration of antiLy6G antibody have been previously reported. In this study, we hypothesized that, the combination of PMF treatments and antiLy6G administration may synergistically potentiate their antiinflammatory actions. The effects of the combination of PMF treatments and antiLy6G administration were investigated by examining the inflammatory signs, histopathological properties of the inflamed site, and measuring the macrophage inflammatory protein-1 alpha (MIP-1α/CCL3) and myeloperoxidase (MPO) levels of inflamed paw tissues in rats with carrageenan-induced acute paw inflammation. In this present study, PMF treatments alone or administration of antiLy6G alone ameliorated the acute inflammation. However, their combination exacerbated the inflammatory signs, hyperalgesia, allodynia, edema and fever, and aggravated the inflammatory conditions by excessive infiltration of inflammatory cells to the inflamed site. These opposing effects of the combined treatments may correlate with enhanced levels of MIP-1α and MPO in inflamed paws. Present results indicated that the combination of the PMF treatments and antiLy6G administration may not provide additional benefits and may actually cause an aggravation of the acute inflammatory process. Findings may also suggest that during neutrophil or immune cell-targeted treatments for inflammatory states, magnetic field exposure may cause unexpected negative consequences.
Collapse
Affiliation(s)
- Tufan Mert
- Department of Biophysics, Faculty of Medicine, Bolu Abant Izzet Baysal University, 14030, Bolu, Turkey.
| | - Tuba Ozcan Metin
- Department of Histology and Embryology, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Mehmet Sahin
- Department of Medical Biology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Selma Yaman
- Department of Biophysics, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| |
Collapse
|
31
|
Depletion of Alveolar Macrophages Increases Pulmonary Neutrophil Infiltration, Tissue Damage, and Sepsis in a Murine Model of Acinetobacter baumannii Pneumonia. Infect Immun 2020; 88:IAI.00128-20. [PMID: 32366576 DOI: 10.1128/iai.00128-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Acinetobacter baumannii has emerged as an important etiological agent of hospital-related infections, especially nosocomial pneumonia. The virulence factors of this bacterium and their interactions with the cells and molecules of the immune system just recently began to be extensively studied. Here, we investigated the impact of alveolar macrophages on A. baumannii pneumonia using a mouse model of infection and a flexible tissue culture system. We hypothesized that depletion of macrophages would enhance sepsis and severity of A. baumannii disease. We showed that macrophages are important for modulating the antibacterial function of neutrophils and play an important role in eradicating A. baumannii infection in vivo Our findings suggest that in the absence of macrophages in the lungs, A. baumannii replicates significantly, and host proinflammatory cytokines are considerably reduced. Neutrophils are abundantly recruited to pulmonary tissue, releasing high amounts of reactive oxygen species and causing extensive tissue damage. The ability of A. baumannii to form biofilms and resist oxidative stress in the respiratory tract facilitates systemic dissemination and ultimately death of infected C57BL/6 mice. These results provide novel information regarding A. baumannii pathogenesis and may be important for the development of therapies aimed at reducing morbidity and mortality associated with this emerging bacterial pathogen.
Collapse
|
32
|
Rambach G, Fleischer V, Harpf V, Lackner M, Meinitzer A, Maier H, Engesser J, Lass-Flörl C, Speth C. Comparative immunopathogenesis in a murine model of inhalative infection with the mucormycetes Lichtheimia corymbifera and Rhizopus arrhizus. PLoS One 2020; 15:e0234063. [PMID: 32555589 PMCID: PMC7299637 DOI: 10.1371/journal.pone.0234063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/18/2020] [Indexed: 12/27/2022] Open
Abstract
Pathogenic mucormycetes induce diseases with considerable morbidity and mortality in immunocompromised patients. Virulence data comparing different Mucorales species and various underlying risk factors are limited. We therefore compared the pathogenesis of inhalative infection by Rhizopus (R.) arrhizus and Lichtheimia (L.) corymbifera in murine models for predominant risk factors for onset of infection. Mice with diabetes or treated with cyclophosphamide or cortisone acetate were challenged via the intranasal route with an isolate of R. arrhizus or L. corymbifera, respectively. Clinical, immunological and inflammation parameters as well as efficacy of posaconazole prophylaxis were monitored over 14 days. Whereas immunocompetent mice showed no clinical symptoms after mucormycete infection, mice treated with either cyclophosphamide (CP) or cortisone acetate (CA) were highly susceptible. Animals infected with the isolate of R. arrhizus showed prolonged survival and lower mortality, compared to those exposed to the L. corymbifera isolate. This lower virulence of R. arrhizus was risk factor-dependent, since diabetic mice died only after infection with Rhizopus, whereas all Lichtheimia-infected diabetic animals survived. Under posaconazole prophylaxis, both mucormycetes were able to establish breakthrough infections in CA- and CP-treated mice, but the course of infection was significantly delayed. Detailed analysis revealed that susceptibility of CA- and CP-treated mice could not be mimicked by exclusive lack or downmodulation of neutrophils, platelets or complement, but can be supposed to be the consequence of a broad immunosuppressive effect induced by the drugs. Both Lichtheimia corymbifera and Rhizopus arrhizus induce invasive mycoses in immunocompromised hosts after inhalative infection. Key parameters such as virulence and immunopathogenesis vary strongly according to fungal species and underlying risk group. Selected neutropenia is no sufficient risk factor for onset of inhalative mucormycosis.
Collapse
Affiliation(s)
- Günter Rambach
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Verena Fleischer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Verena Harpf
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hans Maier
- INNPATH GmbH–Institute of Pathology, Innsbruck, Austria
| | - Johannes Engesser
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
| | - Cornelia Speth
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Invasive Fungal Infections, Innsbruck, Austria
- * E-mail:
| |
Collapse
|
33
|
Abstract
Neutrophils are an essential part of the innate immune system. To study their importance, experimental studies often aim to deplete these cells, generally by injecting anti-Ly6G or anti-Gr1 antibodies. However, these approaches are only partially effective, transient or lack specificity. Here we report that neutrophils remaining after anti-Ly6G treatment are newly derived from the bone marrow, instead of depletion escapees. Mechanistically, newly generated, circulating neutrophils have lower Ly6G membrane expression, and consequently reduced targets for anti-Ly6G-mediated depletion. To overcome this limitation, we develop a double antibody-based depletion strategy that enhances neutrophil elimination by anti-Ly6G treatment. This approach achieves specific, durable and controlled reduction of neutrophils in vivo, and may be suitable for studying neutrophil function in experimental models. Anti-Ly6G or ant-Gr1 antibodies are commonly used to deplete neutrophils in vivo. Here the authors provide mechanistic insight into why these approaches may not specifically or durably reduce the number of neutrophils in mice, and also present a new method that overcomes these limitations to have potentially wide applicability in experimental studies.
Collapse
|
34
|
Mai N, Prifti V, Kim M, Halterman MW. Characterization of neutrophil-neuronal co-cultures to investigate mechanisms of post-ischemic immune-mediated neurotoxicity. J Neurosci Methods 2020; 341:108782. [PMID: 32445795 DOI: 10.1016/j.jneumeth.2020.108782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/23/2020] [Accepted: 05/12/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Immune-mediated reperfusion injury is a critical component of post-ischemic central nervous system (CNS) damage. In this context, the activation and recruitment of polymorphonuclear neutrophils (PMNs) to the CNS induces neurotoxicity in part through the release of degradative enzymes, cytokines, and reactive oxygen species. However, the extent to which close-range interactions between PMNs and neurons contribute to injury in this context has not been directly investigated. NEW METHOD We devised a co-culture model to investigate mechanisms of PMN-dependent neurotoxicity. Specifically, we established the effect of PMN dose, co-incident neuronal ischemia, lipopolysaccharide (LPS)-induced PMN priming, and the requirement for cell-cell contact on cumulative neuron damage. RESULTS AND COMPARISON TO EXISTING METHOD(S) Pre-exposure of day in vitro 10 primary cortical neurons to oxygen-glucose deprivation (OGD) enhanced PMN-dependent neuronal death. Likewise, LPS-induced priming of the PMN donor further increased PMN-induced toxicity in vitro compared to saline-injected controls. Compartmentalization of LPS-primed PMNs using net wells confirmed the requirement for close-range cell-cell interactions in the process of PMN-induced neuronal injury. Moreover, time-lapse imaging and quantitative neurite analyses implicate PMN-neurite interactions in this pathological response. These experiments establish a platform to investigate immune and neural factors that contribute to post-ischemic neurodegeneration. CONCLUSIONS Ischemic and immune priming enhance neurotoxicity in PMN-neuronal co-cultures. Moreover, cell-cell contact and neurite destruction are prominent features in the observed mechanism of post-ischemic neuronal death.
Collapse
Affiliation(s)
- Nguyen Mai
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| | - Viollandi Prifti
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| | - Minsoo Kim
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| | - Marc W Halterman
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States; Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, United States.
| |
Collapse
|
35
|
Burfeind KG, Zhu X, Norgard MA, Levasseur PR, Huisman C, Buenafe AC, Olson B, Michaelis KA, Torres ERS, Jeng S, McWeeney S, Raber J, Marks DL. Circulating myeloid cells invade the central nervous system to mediate cachexia during pancreatic cancer. eLife 2020; 9:e54095. [PMID: 32391790 PMCID: PMC7253193 DOI: 10.7554/elife.54095] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Weight loss and anorexia are common symptoms in cancer patients that occur prior to initiation of cancer therapy. Inflammation in the brain is a driver of these symptoms, yet cellular sources of neuroinflammation during malignancy are unknown. In a mouse model of pancreatic ductal adenocarcinoma (PDAC), we observed early and robust myeloid cell infiltration into the brain. Infiltrating immune cells were predominately neutrophils, which accumulated at a unique central nervous system entry portal called the velum interpositum, where they expressed CCR2. Pharmacologic CCR2 blockade and genetic deletion of Ccr2 both resulted in significantly decreased brain-infiltrating myeloid cells as well as attenuated cachexia during PDAC. Lastly, intracerebroventricular blockade of the purinergic receptor P2RX7 during PDAC abolished immune cell recruitment to the brain and attenuated anorexia. Our data demonstrate a novel function for the CCR2/CCL2 axis in recruiting neutrophils to the brain, which drives anorexia and muscle catabolism.
Collapse
Affiliation(s)
- Kevin G Burfeind
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
- Medical Scientist Training Program, Oregon Health & Science UniversityPortlandUnited States
| | - Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Christian Huisman
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Abigail C Buenafe
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
- Medical Scientist Training Program, Oregon Health & Science UniversityPortlandUnited States
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
- Medical Scientist Training Program, Oregon Health & Science UniversityPortlandUnited States
| | - Eileen RS Torres
- Department of Behavioral Neuroscience, Oregon Health & Science UniversityPortlandUnited States
| | - Sophia Jeng
- Oregon Clinical and Translational Research Institute, Oregon Health & Science UniversityPortlandUnited States
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Shannon McWeeney
- Oregon Clinical and Translational Research Institute, Oregon Health & Science UniversityPortlandUnited States
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandUnited States
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science UniversityPortlandUnited States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science UniversityPortlandUnited States
- Departments of Neurology and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health and & Science UniversityPortlandUnited States
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science UniversityPortlandUnited States
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandUnited States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and & Science University PortlandPortlandUnited States
| |
Collapse
|
36
|
Cleary SJ, Hobbs C, Amison RT, Arnold S, O'Shaughnessy BG, Lefrançais E, Mallavia B, Looney MR, Page CP, Pitchford SC. LPS-induced Lung Platelet Recruitment Occurs Independently from Neutrophils, PSGL-1, and P-Selectin. Am J Respir Cell Mol Biol 2020; 61:232-243. [PMID: 30768917 DOI: 10.1165/rcmb.2018-0182oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Platelets are recruited to inflammatory foci and contribute to host defense and inflammatory responses. Compared with platelet recruitment in hemostasis and thrombosis, the mechanisms of platelet recruitment in inflammation and host defense are poorly understood. Neutrophil recruitment to lung airspaces after inhalation of bacterial LPS requires platelets and PSGL-1 in mice. Given this association between platelets and neutrophils, we investigated whether recruitment of platelets to lungs of mice after LPS inhalation was dependent on PSGL-1, P-selectin, or interaction with neutrophils. BALB/c mice were administered intranasal LPS (O55:B5, 5 mg/kg) and, 48 hours later, lungs were collected and platelets and neutrophils quantified in tissue sections by immunohistochemistry. The effects of functional blocking antibody treatments targeting the platelet-neutrophil adhesion molecules, P-selectin or PSGL-1, or treatment with a neutrophil-depleting antibody targeting Ly6G, were tested on the extent of LPS-induced lung platelet recruitment. Separately in Pf4-Cre × mTmG mice, two-photon intravital microscopy was used to image platelet adhesion in live lungs. Inhalation of LPS caused both platelet and neutrophil recruitment to the lung vasculature. However, decreasing lung neutrophil recruitment by blocking PSGL-1, P-selectin, or depleting blood neutrophils had no effect on lung platelet recruitment. Lung intravital imaging revealed increased adhesion of platelets in the lung microvasculature which was not associated with thrombus formation. In conclusion, platelet recruitment to lungs in response to LPS occurs through mechanisms distinct from those mediating neutrophil recruitment, or the occurrence of pulmonary emboli.
Collapse
Affiliation(s)
- Simon J Cleary
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Carl Hobbs
- 2the Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom; and
| | - Richard T Amison
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Stephanie Arnold
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Blaze G O'Shaughnessy
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Emma Lefrançais
- 3Department of Medicine, University of California San Francisco, San Francisco, California
| | - Beñat Mallavia
- 3Department of Medicine, University of California San Francisco, San Francisco, California
| | - Mark R Looney
- 3Department of Medicine, University of California San Francisco, San Francisco, California
| | - Clive P Page
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Simon C Pitchford
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| |
Collapse
|
37
|
Sun D, Sun P, He S, Shi M. Rat IgG mediated circulatory cell depletion in mice requires mononuclear phagocyte system and is facilitated by complement. J Leukoc Biol 2020; 107:529-539. [PMID: 31965640 DOI: 10.1002/jlb.4a1219-078r] [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: 03/05/2019] [Revised: 12/25/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Application of exogenous Abs targeting cell surface Ags has been widely used as an experimental approach to induce cell depletion or to inhibit receptor functionality. Moreover, Ab therapy is emerging as one of the mainstream strategies for cancer treatment. Previous studies on the mechanisms of Ab-mediated cell depletion mainly employed Abs from the same species as the research subject. However, there has been a recent trend toward using xenogeneic (cross-species) Abs to achieve cell depletion or block receptor-ligand interactions; with rat Abs used in mice being the most common approach. Considering the molecular differences in Abs from different species, the mechanism(s) of xenogeneic Ab-mediated cell depletion is likely to be different than species-matched Ab supplementation. The current work describes our efforts to identify the mechanism of rat anti-mouse Ly6G (clone: 1A8) mAb mediated depletion of mouse neutrophils. The results showed that neutrophils circulating in the blood but not those in the bone marrow are depleted, and depletion depends on mononuclear phagocyte system, especially liver Kupffer cells that efficiently capture and phagocytize targeted cells. Interestingly, whereas species-matched Ab depletion does not require complement functionality, we found that complement activation significantly facilitates cross-species neutrophil depletion. Finally, we found that some rat mAbs (anti-C5aR, anti-CD11a, anti-CD11b, and anti-VLA4) used to block cell surface receptors also induce cell depletion. Thus, our work strongly recommends controlling for cell depletion effect when using these Abs for receptor blockade purposes.
Collapse
Affiliation(s)
- Donglei Sun
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Peng Sun
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA.,School of Agriculture, Ningxia University, Yinchuan, Ningxia, China
| | - Shenghu He
- School of Agriculture, Ningxia University, Yinchuan, Ningxia, China
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
38
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
39
|
Roy-O’Reilly MA, Ahnstedt H, Spychala MS, Munshi Y, Aronowski J, Sansing LH, McCullough LD. Aging exacerbates neutrophil pathogenicity in ischemic stroke. Aging (Albany NY) 2020; 12:436-461. [PMID: 31927534 PMCID: PMC6977697 DOI: 10.18632/aging.102632] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 12/23/2019] [Indexed: 05/28/2023]
Abstract
Ischemic stroke is major cause of disability and mortality worldwide, and aging is strong risk factor for poor post-stroke outcome. Neutrophils traffic rapidly to the brain following ischemic stroke, and recent evidence has suggested that aging may alter neutrophil function after tissue injury. In this study, we hypothesize that aging enhances the pro-inflammatory function of neutrophils, directly contributing to the poorer outcomes seen in aging patients. We utilized demographic data and biological specimens from ischemic stroke patients and an experimental mouse model to determine the correlation between age, neutrophil function and stroke outcomes. In ischemic stroke patients, age was associated with increased mortality and morbidity and higher levels of neutrophil-activating cytokines. In mice, aged animals had higher stroke mortality and morbidity, higher levels of neutrophil-activating cytokines and enhanced generation of neutrophil reactive oxygen species compared to young mice. Finally, depletion of neutrophils via a specific monoclonal antibody after ischemic stroke led to long-term benefits in functional outcome in aged male and female animals, with no benefit observed in young. These results demonstrate that aging is associated with augmented neutrophil pathogenicity in ischemic stroke, and that neutrophil-targeted therapies may confer greater benefit in aged subjects.
Collapse
Affiliation(s)
| | - Hilda Ahnstedt
- Department of Neurology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Monica S. Spychala
- Department of Neurology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Yashasvee Munshi
- Department of Neurology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Jaroslaw Aronowski
- Department of Neurology, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Lauren H. Sansing
- Department of Neurology and Center for Neuroepidemiology and Clinical Neurological Research, Yale School of Medicine, New Haven, CT 06520, USA
| | - Louise D. McCullough
- Department of Neurology, University of Texas Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
40
|
Rosowski EE. Determining macrophage versus neutrophil contributions to innate immunity using larval zebrafish. Dis Model Mech 2020; 13:13/1/dmm041889. [PMID: 31932292 PMCID: PMC6994940 DOI: 10.1242/dmm.041889] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The specific roles of the two major innate immune cell types – neutrophils and macrophages – in response to infection and sterile inflammation are areas of great interest. The larval zebrafish model of innate immunity, and the imaging capabilities it provides, is a source of new research and discoveries in this field. Multiple methods have been developed in larval zebrafish to specifically deplete functional macrophages or neutrophils. Each of these has pros and cons, as well as caveats, that often make it difficult to directly compare results from different studies. The purpose of this Review is to (1) explore the pros, cons and caveats of each of these immune cell-depleted models; (2) highlight and place into a broader context recent key findings on the specific functions of innate immune cells using these models; and (3) explore future directions in which immune cell depletion methods are being expanded. Summary: Macrophages and neutrophils are distinct innate immune cells with diverse roles in diverse inflammatory contexts. Recent research in larval zebrafish using cell-specific depletion methods has revealed new insights into these cells' functions.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
41
|
Liu F, Dai S, Feng D, Peng X, Qin Z, Kearns AC, Huang W, Chen Y, Ergün S, Wang H, Rappaport J, Bryda EC, Chandrasekhar A, Aktas B, Hu H, Chang SL, Gao B, Qin X. Versatile cell ablation tools and their applications to study loss of cell functions. Cell Mol Life Sci 2019; 76:4725-4743. [PMID: 31359086 PMCID: PMC6858955 DOI: 10.1007/s00018-019-03243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Zhongnan Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Yong Chen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, 261053, Weifang, People's Republic of China
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximillan University, 97070, Wurzburg, Germany
| | - Hong Wang
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, 4011 Discovery Drive, Columbia, MO, 65201, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences, 340D Life Sciences Center, University of Missouri, 1201 Rollins St, Columbia, MO, USA
| | - Bertal Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
42
|
Lee HH, Moon Y, Shin JS, Lee JH, Kim TW, Jang C, Park C, Lee J, Kim Y, Kim Y, Werz O, Park BY, Lee JY, Lee KT. A novel mPGES-1 inhibitor alleviates inflammatory responses by downregulating PGE2 in experimental models. Prostaglandins Other Lipid Mediat 2019; 144:106347. [DOI: 10.1016/j.prostaglandins.2019.106347] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
|
43
|
Zhao J, Huangfu C, Chang Z, Grainger AT, Liu Z, Shi W. Atherogenesis in the Carotid Artery with and without Interrupted Blood Flow of Two Hyperlipidemic Mouse Strains. J Vasc Res 2019; 56:241-254. [PMID: 31536996 DOI: 10.1159/000502691] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 08/13/2019] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Atherosclerosis in the carotid arteries is a common cause of ischemic stroke. We examined atherogenesis in the left carotid artery with and without interrupted blood flow of C57BL/6 (B6) and C3H-Apoe-deficient (Apoe-/-) mouse strains. METHODS Blood flow was interrupted by ligating the common carotid artery near its bifurcation in one group of mice and another group was not interrupted. RESULTS Without interference with blood flow, C3H-Apoe-/- mice developed no atherosclerosis in the carotid artery, while B6-Apoe-/- mice formed advanced atherosclerotic lesions (98,019 ± 10,594 μm2/section) after 12 weeks of a Western diet. When blood flow was interrupted by ligating the common carotid artery near its bifurcation, C3H-Apoe-/- mice showed fatty streak lesions 2 weeks after ligation, and by 4 weeks fibrous lesions had formed, although they were smaller than in B6-Apoe-/- mice. Neutrophil adhesion to endothelium and infiltration in lesions was observed in ligated arteries of both strains. Treatment of B6-Apoe-/- mice with antibody against neutrophils had little effect on lesion size. CONCLUSIONS These findings demonstrate the dramatic influences of genetic backgrounds and blood flow on atherogenesis in the carotid artery of hyperlipidemic mice.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chaoji Huangfu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA.,Center for Disease Control and Prevention, Western Theater Command, Lanzhou, China
| | - Zhihui Chang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Andrew T Grainger
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| | - Zhaoyu Liu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA, .,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA,
| |
Collapse
|
44
|
Limitations of neutrophil depletion by anti-Ly6G antibodies in two heterogenic immunological models. Immunol Lett 2019; 212:30-36. [DOI: 10.1016/j.imlet.2019.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/24/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
|
45
|
Neutrophils Dampen Adaptive Immunity in Brucellosis. Infect Immun 2019; 87:IAI.00118-19. [PMID: 30804100 PMCID: PMC6479033 DOI: 10.1128/iai.00118-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
Brucella organisms are intracellular stealth pathogens of animals and humans. The bacteria overcome the assault of innate immunity at early stages of an infection. Brucella organisms are intracellular stealth pathogens of animals and humans. The bacteria overcome the assault of innate immunity at early stages of an infection. Removal of polymorphonuclear neutrophils (PMNs) at the onset of adaptive immunity against Brucella abortus favored bacterial elimination in mice. This was associated with higher levels of interferon gamma (IFN-γ) and a higher proportion of cells expressing interleukin 6 (IL-6) and inducible nitric oxide synthase (iNOS), compatible with M1 macrophages, in PMN-depleted B. abortus-infected (PMNd-Br) mice. At later times in the acute infection phase, the amounts of IFN-γ fell while IL-6, IL-10, and IL-12 became the predominant cytokines in PMNd-Br mice. IL-4, IL-1β, and tumor necrosis factor alpha (TNF-α) remained at background levels at all times of the infection. Depletion of PMNs at the acute stages of infection promoted the premature resolution of spleen inflammation. The efficient removal of bacteria in the PMNd-Br mice was not due to an increase of antibodies, since the immunoglobulin isotype responses to Brucella antigens were dampened. Anti-Brucella antibodies abrogated the production of IL-6, IL-10, and IL-12 but did not affect the levels of IFN-γ at later stages of infection in PMNd-Br mice. These results demonstrate that PMNs have an active role in modulating the course of B. abortus infection after the adaptive immune response has already developed.
Collapse
|
46
|
Jarrot PA, Tellier E, Plantureux L, Crescence L, Robert S, Chareyre C, Daniel L, Secq V, Garcia S, Dignat-George F, Panicot-Dubois L, Dubois C, Kaplanski G. Neutrophil extracellular traps are associated with the pathogenesis of diffuse alveolar hemorrhage in murine lupus. J Autoimmun 2019; 100:120-130. [PMID: 30930069 DOI: 10.1016/j.jaut.2019.03.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/30/2022]
Abstract
Diffuse alveolar hemorrhage (DAH) is a life-threatening complication of systemic lupus erythematosus (SLE) and systemic vasculitis. Although initially described to have antibacterial properties, increasing evidence suggests that neutrophil extracellular traps (NETs) have a detrimental role in both autoimmune diseases and acute lung injury. We investigated whether NETs could be detected in a murine model of pristane-induced lupus DAH and contribute to lung injury. Such NETs might constitute a therapeutic target. NETs were characterized by immunofluorescence staining of DNA, neutrophil elastase and citrullinated histones. Evaluation of lung injury was performed by haematoxylin-eosin staining and a quantification program. Clinical status of the mice was assessed by measurement of arterial oxygen saturation and survival curves after recombinant human deoxyribonuclease-1 (Rh-DNase-1) inhalations or polymorphonuclear neutrophil (PMN) depletion. Pristane was found to promote NETs formation in vitro and in vivo. Treatment of mice with Rh-DNase-1 inhalations cleared NETs and reduced lung injury. Clinical status improved significantly, with increased arterial oxygenation and survival. Following PMN depletion, NETs were absent with a subsequent reduction of lung injury and improved arterial oxygenation. These results support a pathogenic role of PMNs and NETs in lung injury during pristane-induced DAH. Targeting NETs with Rh-DNase-1 inhalations could constitute an interesting adjuvant therapy in human DAH.
Collapse
Affiliation(s)
- Pierre-André Jarrot
- Aix-Marseille Univ, INSERM, INRA, C2VN, Marseille, France; Department of Internal Medicine and Clinical Immunology CHU Conception, Assistance Publique-Hôpitaux de Marseille (AP-HM), Marseille, France.
| | - Edwige Tellier
- Aix-Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Lea Plantureux
- Aix-Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | | | | | | | - Laurent Daniel
- Laboratory of Pathology, CHU La Timone, AP-HM, Marseille, France
| | - Véronique Secq
- Laboratory of Pathology, CHU Nord, AP-HM, Marseille, France
| | | | - Françoise Dignat-George
- Aix-Marseille Univ, INSERM, INRA, C2VN, Marseille, France; Laboratory of Haematology, CHU La Conception, AP-HM, Marseille, France
| | | | | | - Gilles Kaplanski
- Aix-Marseille Univ, INSERM, INRA, C2VN, Marseille, France; Department of Internal Medicine and Clinical Immunology CHU Conception, Assistance Publique-Hôpitaux de Marseille (AP-HM), Marseille, France
| |
Collapse
|
47
|
Anti-inflammatory properties of Liposome-encapsulated clodronate or Anti-Ly6G can be modulated by peripheral or central inflammatory markers in carrageenan-induced inflammation model. Inflammopharmacology 2019; 27:603-612. [DOI: 10.1007/s10787-019-00563-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/03/2019] [Indexed: 01/13/2023]
|
48
|
Carrasco-Yepez MM, Contis-Montes de Oca A, Campos-Rodriguez R, Falcon-Acosta D, Pacheco-Yepez J, Rodriguez-Mera IB, Bonilla-Lemus P, Rosales-Cruz E, Lopez-Reyes I, Rojas-Hernandez S. Mouse neutrophils release extracellular traps in response to Naegleria fowleri. Parasite Immunol 2019; 41:e12610. [PMID: 30525201 DOI: 10.1111/pim.12610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/27/2018] [Indexed: 01/09/2023]
Abstract
Naegleria fowleri is a free-living amoeba, which is able to infect humans through the nasal mucosa causing a disease in the central nervous system known as primary amoebic meningoencephalitis (PAM). Polymorphonuclear cells (PMNs) play a critical role in the early phase of N fowleri infection. Recently, a new biological defence mechanism called neutrophil extracellular traps (NETs) has been attracting attention. These structures represent an important strategy to immobilize and kill invading microorganisms. In this work, we evaluate the capacity of N fowleri to induce the NETs release by PMNs cells in mice in vitro and in vivo. In vitro: Neutrophils from bone marrow were cocultured with N fowleri trophozoites. In vivo: we employed a mouse model of PAM. We evaluated DNA, histone and myeloperoxidase (MPO) and the formation of NETs by confocal microscopy. Our results showed N fowleri induce both NETs and MPO release by PMNs cells in mice after trophozoite exposure, which increased through time, in vitro and in vivo. These results demonstrate that NETs are somehow associated with the amoebas. We suggest PMNs release their traps trying to avoid N fowleri attachment at the apical side of the nasal epithelium.
Collapse
Affiliation(s)
| | - Arturo Contis-Montes de Oca
- Laboratorio de Inmunobiología Molecular y Celular, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| | - Rafael Campos-Rodriguez
- Laboratorio de Bioquímica, Departamento de Bioquímica, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| | - Diana Falcon-Acosta
- Laboratorio de Inmunobiología Molecular y Celular, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| | - Judith Pacheco-Yepez
- Laboratorio de Bioquímica, Departamento de Bioquímica, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| | - Itzel Berenice Rodriguez-Mera
- Laboratorio de Inmunobiología Molecular y Celular, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| | - Patricia Bonilla-Lemus
- Laboratorio de Microbiología, Proyecto CyMA, UIICSE, UNAM FES Iztacala, Tlalnepantla, México
| | - Erika Rosales-Cruz
- Laboratorio de Investigación en Hematopatología, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, México City, México
| | - Israel Lopez-Reyes
- Universidad de la Ciudad de México, Plantel Cuautepec, Mexico City, Mexico
| | - Saul Rojas-Hernandez
- Laboratorio de Inmunobiología Molecular y Celular, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| |
Collapse
|
49
|
Mendez J, Sun D, Tuo W, Xiao Z. Bovine neutrophils form extracellular traps in response to the gastrointestinal parasite Ostertagia ostertagi. Sci Rep 2018; 8:17598. [PMID: 30514873 PMCID: PMC6279769 DOI: 10.1038/s41598-018-36070-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022] Open
Abstract
Ostertagia ostertagi (OO) is a widespread parasite that causes chronic infection in cattle and leads to annual losses of billions of dollars in the cattle industry. It remains unclear why cattle are unable to mount an effective immune response despite a large influx of immune cells to the infected abomasal mucosa and draining lymph nodes. Neutrophils, the immune system’s first responders, have the capacity to release neutrophil extracellular traps (NETs) to contain various pathogens, including some parasites. In the present study, the mechanisms by which O. ostertagi influences bovine NET formation were investigated. O. ostertagi larval soluble extract (OO extract) was able to induce typical NETs by purified neutrophils in vitro, confirmed by co-localization of extracellular DNA with typical NET-associated proteins histone and neutrophil elastase (NE). Consistent with existing literature, inhibition assays demonstrated that these OO extract-induced NETs were dependent upon the enzymes NADPH oxidase and myeloperoxidase (MPO). Live OO stage 4 larvae (L4) stimulated neutrophils to form NETs similar to those induced by OO extract. Bovine neutrophils also released NETs in response to Caenorhabditis elegans, a free-living soil nematode, suggesting that bovine NET production may be a conserved mechanism against a broad range of nematodes. This is the first report demonstrating O. ostertagi-induced NET formation by bovine neutrophils, a potentially underappreciated mechanism in the early immune response against nematode infections.
Collapse
Affiliation(s)
- Jonatan Mendez
- Department of Avian and Animal Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Donglei Sun
- Department of Veterinary Medicine, University of Maryland, College Park, MD, 20742, USA
| | - Wenbin Tuo
- Animal Parasitic Diseases Laboratory, USDA/ARS, Beltsville, MD, 20705, USA.
| | - Zhengguo Xiao
- Department of Avian and Animal Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
50
|
Prostaglandin E2 produced by myeloid-derived suppressive cells induces cancer stem cells in uterine cervical cancer. Oncotarget 2018; 9:36317-36330. [PMID: 30555631 PMCID: PMC6284736 DOI: 10.18632/oncotarget.26347] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/24/2018] [Indexed: 11/25/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) enhance tumor progression by suppressing tumor-specific T cell responses, stimulating tumor angiogenesis, or promoting tumor cell metastasis. However, the biology of MDSCs have not been fully investigated. In the current study, we investigated the role of MDSCs in inducing cancer stem-like cells and explored a clinically feasible approach for targeting MDSCs-mediated cancer stem-like cells induction. In vitro and in vivo experiments revealed that MDSCs induced by tumor-derived G-CSF enhanced the stemness of cervical cancer cells by producing Prostaglandin E2 (PGE2). We also demonstrated that anti-Gr-1 neutralizing antibody or celecoxib inhibited the induction of cancer stem-like cells and enhanced the efficacy of cisplatin in cervical cancer. In clinical samples, MDSCs, PGE2, and CSCs had positive correlations. In conclusion, G-CSF-induced MDSCs enhance the stemness of uterine cervical cancer cells by producing PGE2. Targeting MDSCs or PGE2 might be a reasonable strategy for enhancing the efficacies of treatments.
Collapse
|