151
|
Lu H, Lin J, Xu C, Sun M, Zuo K, Zhang X, Li M, Huang H, Li Z, Wu W, Feng B, Liu Z. Cyclosporine modulates neutrophil functions via the SIRT6-HIF-1α-glycolysis axis to alleviate severe ulcerative colitis. Clin Transl Med 2021; 11:e334. [PMID: 33634990 PMCID: PMC7882115 DOI: 10.1002/ctm2.334] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cyclosporine A (CsA) is routinely used to treat patients with steroid-refractory acute severe ulcerative colitis (ASUC). Here, we studied the underlying mechanisms of CsA-mediated alleviation in ASUC patients. METHODS Neutrophil functions including expression of cytokines, apoptosis, and migration were measured by qRT-PCR, flow cytometry, and Transwell assay. Dynamic changes of glycolysis and tricarboxylic acid (TCA) cycle were measured by a Seahorse extracellular flux analyzer. Gene differences were determined and verified by RNA sequencing, qRT-PCR, and Western blotting. Small interfering RNA and inhibitors were used to knock down Sirtuin 6 (SIRT6) in HL-60 cells and block expression of SIRT6, hypoxia-inducible factor-1α (HIF-1α), and pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4) in neutrophils. RESULTS We found that HIF-1α expression and glycolysis significantly increased, while the release of IL-8, myeloperoxidase (MPO) and reactive oxygen species (ROS), the apoptosis, and ability of migration markedly decreased in neutrophils of ASUC patients who responded to CsA (Response group) compared with those who did not respond to CsA (Nonresponse group). We also observed that CsA-induced functional alternation of neutrophils was initiated through suppressing SIRT6 expression, which is responsible for expression of the downstream signaling molecules (e.g., HIF-1α, PFKFB3) and PDK4 ubiquitination, leading to fueling neutrophil glycolysis and TCA cycle. Furthermore, blockage of SIRT6 signaling demonstrated to be the same functional changes as CsA to decrease the migration of neutrophils. CONCLUSIONS The data reveal a novel mechanism of CsA in alleviating ASUC by promoting neutrophil HIF-1α expression and restricting excessive neutrophil activation in a SIRT6-HIF-1α-glycolysis axis, suggesting SIRT6 as a candidate target for maintaining mucosal homeostasis and treating intestinal inflammation.
Collapse
Affiliation(s)
- Huiying Lu
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
| | - Jian Lin
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
| | - Chunjin Xu
- Department of GastroenterologyFirst People's Hospital of Shangqiu City Affiliated to Xinxiang Medical UniversityShangqiuChina
| | - Mingming Sun
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
| | - Keqiang Zuo
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
| | - Xiaoping Zhang
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
| | - Mingsong Li
- Department of GastroenterologyThird Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Hailiang Huang
- Analytic and Translational Genetics UnitMassachusetts General HospitalBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
- Broad Institute of Harvard and MITCambridgeMassachusettsUSA
| | - Zhong Li
- Shanghai Cell Therapy GroupShanghaiChina
| | - Wei Wu
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
| | - Baisui Feng
- Department of GastroenterologySecond Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhanju Liu
- Center for IBD ResearchDepartment of GastroenterologyShanghai Tenth People's Hospital of Tongji UniversityShanghaiChina
- Department of GastroenterologySecond Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
152
|
Spijkerman R, Jorritsma NKN, Bongers SH, Bindels BJJ, Jukema BN, Hesselink L, Hietbrink F, Leenen LPH, van Goor HMR, Vrisekoop N, Kaasjager KAH, Koenderman L. An increase in CD62L dim neutrophils precedes the development of pulmonary embolisms in COVID-19 patients. Scand J Immunol 2021; 93:e13023. [PMID: 33482019 PMCID: PMC7995011 DOI: 10.1111/sji.13023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/06/2020] [Accepted: 01/17/2021] [Indexed: 11/30/2022]
Abstract
Objectives A high incidence of pulmonary embolism (PE) is reported in patients with critical coronavirus disease 2019 (COVID‐19). Neutrophils may contribute to this through a process referred to as immunothrombosis. The aim of this study was to investigate the occurrence of neutrophil subpopulations in blood preceding the development of COVID‐19 associated PE. Methods We studied COVID‐19 patients admitted to the ICU of our tertiary hospital between 19‐03‐2020 and 17‐05‐2020. Point‐of‐care fully automated flow cytometry was performed prior to ICU admission, measuring the neutrophil activation/maturation markers CD10, CD11b, CD16 and CD62L. Neutrophil receptor expression was compared between patients who did or did not develop PE (as diagnosed on CT angiography) during or after their ICU stay. Results Among 25 eligible ICU patients, 22 subjects were included for analysis, of whom nine developed PE. The median (IQR) time between neutrophil phenotyping and PE occurrence was 9 (7‐12) days. A significant increase in the immune‐suppressive neutrophil phenotype CD16bright/CD62Ldim was observed on the day of ICU admission (P = 0.014) in patients developing PE compared to patients who did not. Conclusion The increase in this neutrophil phenotype indicates that the increased number of CD16bright/CD62Ldim neutrophils might be used as prognostic marker to predict those patients that will develop PE in critical COVID‐19 patients.
Collapse
Affiliation(s)
- Roy Spijkerman
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nikita K N Jorritsma
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Suzanne H Bongers
- Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bas J J Bindels
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bernard N Jukema
- Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lillian Hesselink
- Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Falco Hietbrink
- Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Luke P H Leenen
- Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Harriët M R van Goor
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nienke Vrisekoop
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karin A H Kaasjager
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
153
|
Swartzwelter BJ, Verde A, Rehak L, Madej M, Puntes VF, De Luca AC, Boraschi D, Italiani P. Interaction between Macrophages and Nanoparticles: In Vitro 3D Cultures for the Realistic Assessment of Inflammatory Activation and Modulation of Innate Memory. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:207. [PMID: 33467414 PMCID: PMC7830034 DOI: 10.3390/nano11010207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
Understanding the modes of interaction between human monocytes/macrophages and engineered nanoparticles is the basis for assessing particle safety, in terms of activation of innate/inflammatory reactions, and their possible exploitation for medical applications. In vitro assessment of nanoparticle-macrophage interaction allows for examining the response of primary human cells, but the conventional 2D cultures do not reproduce the three-dimensional spacing of a tissue and the interaction of macrophages with the extracellular tissue matrix, conditions that shape macrophage recognition capacity and reactivity. Here, we have compared traditional 2D cultures with cultures on a 3D collagen matrix for evaluating the capacity gold nanoparticles to induce monocyte activation and subsequent innate memory in human blood monocytes in comparison to bacterial LPS. Results show that monocytes react to stimuli almost in the same way in 2D and 3D cultures in terms of production of TNFα and IL-6, but that notable differences are found when IL-8 and IL-1Ra are examined, in particular in the recall/memory response of primed cells to a second stimulation, with the 3D cultures showing cell activation and memory effects of nanoparticles better. In addition, the response variations in monocytes/macrophages from different donors point towards a personalized assessment of the nanoparticle effects on macrophage activation.
Collapse
Affiliation(s)
- Benjamin J. Swartzwelter
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Alessandro Verde
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Laura Rehak
- Athena Biomedical Innovations, 00100 Roma, Italy;
| | - Mariusz Madej
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Victor. F. Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and The Barcelona Institute of Science and Technology (BIST), Campus UAB, 08193 Bellaterra, Barcelona, Spain;
| | - Anna Chiara De Luca
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
154
|
Lou X, Chen Z, He Z, Sun M, Sun J. Bacteria-Mediated Synergistic Cancer Therapy: Small Microbiome Has a Big Hope. NANO-MICRO LETTERS 2021; 13:37. [PMID: 34138211 PMCID: PMC8187705 DOI: 10.1007/s40820-020-00560-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/22/2020] [Indexed: 05/03/2023]
Abstract
The use of bacteria to specifically migrate to cancerous tissue and elicit an antitumor immune response provides a promising platform against cancer with significantly high potency. With dozens of clinical trials underway, some researchers hold the following views: "humans are nearing the first commercial live bacteria therapeutic." However, the facultative anaerobe Salmonella typhimurium VNP20009, which is particularly safe and shows anticancer effects in preclinical studies, had failed in a phase I clinical trial due to low tumor regression and undesired dose-dependent side effects. This is almost certain to disappoint people's inflated expectations, but it is noted that recent state-of-the-art research has turned attention to bacteria-mediated synergistic cancer therapy (BMSCT). In this review, the foundation of bacteria-mediated bio-therapy is outlined. Then, we summarize the potential benefits and challenges of bacterial bio-therapy in combination with different traditional anticancer therapeutic modalities (chemotherapy, photothermal therapy, reactive oxygen and nitrogen species therapy, immunotherapy, or prodrug-activating therapy) in the past 5 years. Next, we discuss multiple administration routes of BMSCT, highlighting potentiated antitumor responses and avoidance of potential side effects. Finally, we envision the opportunities and challenges for BMSCT development, with the purpose of inspiring medicinal scientists to widely utilize the microbiome approach in patient populations.
Collapse
Affiliation(s)
- Xinyu Lou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Zhichao Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Mengchi Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China.
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|
155
|
Ulfig A, Leichert LI. The effects of neutrophil-generated hypochlorous acid and other hypohalous acids on host and pathogens. Cell Mol Life Sci 2021; 78:385-414. [PMID: 32661559 PMCID: PMC7873122 DOI: 10.1007/s00018-020-03591-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Neutrophils are predominant immune cells that protect the human body against infections by deploying sophisticated antimicrobial strategies including phagocytosis of bacteria and neutrophil extracellular trap (NET) formation. Here, we provide an overview of the mechanisms by which neutrophils kill exogenous pathogens before we focus on one particular weapon in their arsenal: the generation of the oxidizing hypohalous acids HOCl, HOBr and HOSCN during the so-called oxidative burst by the enzyme myeloperoxidase. We look at the effects of these hypohalous acids on biological systems in general and proteins in particular and turn our attention to bacterial strategies to survive HOCl stress. HOCl is a strong inducer of protein aggregation, which bacteria can counteract by chaperone-like holdases that bind unfolding proteins without the need for energy in the form of ATP. These chaperones are activated by HOCl through thiol oxidation (Hsp33) or N-chlorination of basic amino acid side-chains (RidA and CnoX) and contribute to bacterial survival during HOCl stress. However, neutrophil-generated hypohalous acids also affect the host system. Recent studies have shown that plasma proteins act not only as sinks for HOCl, but get actively transformed into modulators of the cellular immune response through N-chlorination. N-chlorinated serum albumin can prevent aggregation of proteins, stimulate immune cells, and act as a pro-survival factor for immune cells in the presence of cytotoxic antigens. Finally, we take a look at the emerging role of HOCl as a potential signaling molecule, particularly its role in neutrophil extracellular trap formation.
Collapse
Affiliation(s)
- Agnes Ulfig
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Lars I Leichert
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
156
|
Guo Q, Zhao Y, Li J, Liu J, Yang X, Guo X, Kuang M, Xia H, Zhang Z, Cao L, Luo Y, Bao L, Wang X, Wei X, Deng W, Wang N, Chen L, Chen J, Zhu H, Gao R, Qin C, Wang X, You F. Induction of alarmin S100A8/A9 mediates activation of aberrant neutrophils in the pathogenesis of COVID-19. Cell Host Microbe 2020; 29:222-235.e4. [PMID: 33388094 PMCID: PMC7762710 DOI: 10.1016/j.chom.2020.12.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/11/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic poses an unprecedented public health crisis. Evidence suggests that SARS-CoV-2 infection causes dysregulation of the immune system. However, the unique signature of early immune responses remains elusive. We characterized the transcriptome of rhesus macaques and mice infected with SARS-CoV-2. Alarmin S100A8 was robustly induced in SARS-CoV-2-infected animal models as well as in COVID-19 patients. Paquinimod, a specific inhibitor of S100A8/A9, could rescue the pneumonia with substantial reduction of viral loads in SARS-CoV-2-infected mice. Remarkably, Paquinimod treatment resulted in almost 100% survival in a lethal model of mouse coronavirus infection using the mouse hepatitis virus (MHV). A group of neutrophils that contributes to the uncontrolled pathological damage and onset of COVID-19 was dramatically induced by coronavirus infection. Paquinimod treatment could reduce these neutrophils and regain anti-viral responses, unveiling key roles of S100A8/A9 and aberrant neutrophils in the pathogenesis of COVID-19, highlighting new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Qirui Guo
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Yingchi Zhao
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Junhong Li
- University of Chinese Academy of Sciences, CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jiangning Liu
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xiuhong Yang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xuefei Guo
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Ming Kuang
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Huawei Xia
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Zeming Zhang
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Lili Cao
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Yujie Luo
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Linlin Bao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xiao Wang
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Xuemei Wei
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Wei Deng
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Nan Wang
- University of Chinese Academy of Sciences, CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Luoying Chen
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Jingxuan Chen
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China
| | - Hua Zhu
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Ran Gao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Chuan Qin
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.
| | - Xiangxi Wang
- University of Chinese Academy of Sciences, CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Fuping You
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
157
|
Feng Y, Zhang N, Wang S, Zou W, He Y, Ma JA, Liu P, Liu X, Hu C, Hou T. Systemic Inflammation Response Index Is a Predictor of Poor Survival in Locally Advanced Nasopharyngeal Carcinoma: A Propensity Score Matching Study. Front Oncol 2020; 10:575417. [PMID: 33363009 PMCID: PMC7759154 DOI: 10.3389/fonc.2020.575417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Nasopharyngeal carcinoma (NPC) is a common malignancy in China and known prognostic factors are limited. In this study, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune inflammation index (SII), and systemic inflammation response index (SIRI) were evaluated as prognostic factors in locally advanced NPC patients. Materials and Methods NPC patients who received curative radiation or chemoradiation between January 2012 and December 2015 at the Second Xiangya Hospital were retrospectively reviewed, and a total of 516 patients were shortlisted. After propensity score matching (PSM), 417 patients were eventually enrolled. Laboratory and clinical data were collected from the patients' records. Receiver operating characteristic curve analysis was used to determine the optimal cut-off value. Survival curves were analyzed using the Kaplan-Meier method. The Cox proportional hazard model was used to identify prognostic variables. Results After PSM, all basic characteristics between patients in the high SIRI group and low SIRI group were balanced except for sex (p=0.001) and clinical stage (p=0.036). Univariate analysis showed that NLR (p=0.001), PLR (p=0.008), SII (p=0.001), and SIRI (p<0.001) were prognostic factors for progression-free survival (PFS) and overall survival (OS). However, further multivariate Cox regression analysis showed that only SIRI was an independent predictor of PFS and OS (hazard ratio (HR):2.83; 95% confidence interval (CI): 1.561-5.131; p=0.001, HR: 5.19; 95% CI: 2.588-10.406; p<0.001), respectively. Conclusion Our findings indicate that SIRI might be a promising predictive indicator of locally advanced NPC patients.
Collapse
Affiliation(s)
- Yuhua Feng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sisi Wang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen Zou
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan He
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-An Ma
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ping Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xianling Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunhong Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tao Hou
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
158
|
Ballesteros I, Rubio-Ponce A, Genua M, Lusito E, Kwok I, Fernández-Calvo G, Khoyratty TE, van Grinsven E, González-Hernández S, Nicolás-Ávila JÁ, Vicanolo T, Maccataio A, Benguría A, Li JL, Adrover JM, Aroca-Crevillen A, Quintana JA, Martín-Salamanca S, Mayo F, Ascher S, Barbiera G, Soehnlein O, Gunzer M, Ginhoux F, Sánchez-Cabo F, Nistal-Villán E, Schulz C, Dopazo A, Reinhardt C, Udalova IA, Ng LG, Ostuni R, Hidalgo A. Co-option of Neutrophil Fates by Tissue Environments. Cell 2020; 183:1282-1297.e18. [PMID: 33098771 DOI: 10.1016/j.cell.2020.10.003] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/10/2020] [Accepted: 10/01/2020] [Indexed: 02/09/2023]
Abstract
Classically considered short-lived and purely defensive leukocytes, neutrophils are unique in their fast and moldable response to stimulation. This plastic behavior may underlie variable and even antagonistic functions during inflammation or cancer, yet the full spectrum of neutrophil properties as they enter healthy tissues remains unexplored. Using a new model to track neutrophil fates, we found short but variable lifetimes across multiple tissues. Through analysis of the receptor, transcriptional, and chromatin accessibility landscapes, we identify varying neutrophil states and assign non-canonical functions, including vascular repair and hematopoietic homeostasis. Accordingly, depletion of neutrophils compromised angiogenesis during early age, genotoxic injury, and viral infection, and impaired hematopoietic recovery after irradiation. Neutrophils acquired these properties in target tissues, a process that, in the lungs, occurred in CXCL12-rich areas and relied on CXCR4. Our results reveal that tissues co-opt neutrophils en route for elimination to induce programs that support their physiological demands.
Collapse
Affiliation(s)
- Iván Ballesteros
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain.
| | - Andrea Rubio-Ponce
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain; Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Marco Genua
- Vita-Salute San Raffaele University and San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Eleonora Lusito
- Vita-Salute San Raffaele University and San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Immanuel Kwok
- Singapore Immunology Nework (SIgN), A(∗)STAR, Biopolis, Singapore 138648, Singapore
| | - Gabriel Fernández-Calvo
- Department of Mathematics & MOLAB-Mathematical Oncology Laboratory, University of Castilla-La Mancha, Ciudad Real 13001, Spain
| | - Tariq E Khoyratty
- Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, UK
| | | | - Sara González-Hernández
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - José Ángel Nicolás-Ávila
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Tommaso Vicanolo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Antonio Maccataio
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Alberto Benguría
- Genomic Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Jackson LiangYao Li
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain; Singapore Immunology Nework (SIgN), A(∗)STAR, Biopolis, Singapore 138648, Singapore
| | - José M Adrover
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Alejandra Aroca-Crevillen
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Juan A Quintana
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Sandra Martín-Salamanca
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Francisco Mayo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Stefanie Ascher
- Institute for Pharmacy & Biochemistry, Johannes Gutenberg University of Mainz, Johann-Joachim-Becher-Weg 30, Mainz 55128, Germany
| | - Giulia Barbiera
- Vita-Salute San Raffaele University and San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universitat, Munich 80802, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen 445141, Germany
| | - Florent Ginhoux
- Singapore Immunology Nework (SIgN), A(∗)STAR, Biopolis, Singapore 138648, Singapore
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Estanislao Nistal-Villán
- Microbiology Section, Department Pharmacological and Health Sciences, Facultad de Farmacia, Universidad CEU San Pablo, Madrid 28668, Spain
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, LMU Klinikum, Ludwig-Maximilians-Universität, Munich 80336, Germany; DZHK (German Centre for Cardiovascular Research), Munich 80802, Germany
| | - Ana Dopazo
- Genomic Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis Mainz (CTH), University Medical Center Mainz, Johannes Gutenberg University of Mainz, Mainz 55131, Germany
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, OX3 7FY, UK
| | - Lai Guan Ng
- Singapore Immunology Nework (SIgN), A(∗)STAR, Biopolis, Singapore 138648, Singapore
| | - Renato Ostuni
- Vita-Salute San Raffaele University and San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Andrés Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universitat, Munich 80802, Germany.
| |
Collapse
|
159
|
Deryugina E, Carré A, Ardi V, Muramatsu T, Schmidt J, Pham C, Quigley JP. Neutrophil Elastase Facilitates Tumor Cell Intravasation and Early Metastatic Events. iScience 2020; 23:101799. [PMID: 33299970 PMCID: PMC7702017 DOI: 10.1016/j.isci.2020.101799] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/15/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
Functional roles of neutrophil elastase (NE) have not been examined in distinct steps of the metastatic cascade. NE, delivered to primary tumors as a purified enzyme or within intact neutrophils or neutrophil granule content, enhanced human tumor cell intravasation and subsequent dissemination via NE-mediated formation of dilated intratumoral vasculature. These effects depended on picomole range of NE activity, sensitive to its natural inhibitor, α1PI. In Elane-negative mice, the lack of NE decreased lung retention of human tumor cells in experimental metastasis. Furthermore, NE was essential for spontaneous metastasis of murine carcinoma cells in a syngeneic orthotopic model of oral cancer. NE also induced tumor cell survival and migration via Src/PI3K-dependent activation of Akt signaling, vital for tumor cell dissemination in vivo. Together, our findings implicate NE, a potent host enzyme specific for first-responding innate immune cells, as directly involved in early metastatic events and a potential target for therapeutic intervention. NE enhances human carcinoma cell intravasation and spontaneous metastasis NE mediates formation of dilated intratumoral vasculature supporting cell intravasation NE-KO mice exhibit decreased lung retention and spontaneous metastasis of tumor cells NE induces tumor cell survival and migration via activation of Src/PI3K/Akt pathway
Collapse
Affiliation(s)
- Elena Deryugina
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alexia Carré
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Veronica Ardi
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.,National University, 9388 Lightwave Avenue, San Diego, CA 92123, USA
| | - Tomoki Muramatsu
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jonas Schmidt
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Christine Pham
- Department of Internal Medicine, Washington University, St. Louis, MO 63110, USA
| | - James P Quigley
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
160
|
Moorlag SJCFM, Rodriguez-Rosales YA, Gillard J, Fanucchi S, Theunissen K, Novakovic B, de Bont CM, Negishi Y, Fok ET, Kalafati L, Verginis P, Mourits VP, Koeken VACM, de Bree LCJ, Pruijn GJM, Fenwick C, van Crevel R, Joosten LAB, Joosten I, Koenen H, Mhlanga MM, Diavatopoulos DA, Chavakis T, Netea MG. BCG Vaccination Induces Long-Term Functional Reprogramming of Human Neutrophils. Cell Rep 2020; 33:108387. [PMID: 33207187 PMCID: PMC7672522 DOI: 10.1016/j.celrep.2020.108387] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 07/16/2020] [Accepted: 10/23/2020] [Indexed: 12/31/2022] Open
Abstract
The tuberculosis vaccine bacillus Calmette-Guérin (BCG) protects against some heterologous infections, probably via induction of non-specific innate immune memory in monocytes and natural killer (NK) cells, a process known as trained immunity. Recent studies have revealed that the induction of trained immunity is associated with a bias toward granulopoiesis in bone marrow hematopoietic progenitor cells, but it is unknown whether BCG vaccination also leads to functional reprogramming of mature neutrophils. Here, we show that BCG vaccination of healthy humans induces long-lasting changes in neutrophil phenotype, characterized by increased expression of activation markers and antimicrobial function. The enhanced function of human neutrophils persists for at least 3 months after vaccination and is associated with genome-wide epigenetic modifications in trimethylation at histone 3 lysine 4. Functional reprogramming of neutrophils by the induction of trained immunity might offer novel therapeutic strategies in clinical conditions that could benefit from modulation of neutrophil effector function.
Collapse
Affiliation(s)
- Simone J C F M Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yessica Alina Rodriguez-Rosales
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joshua Gillard
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands; Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, the Netherlands
| | - Stephanie Fanucchi
- Division of Chemical, Systems & Synthetic Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Kate Theunissen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Boris Novakovic
- Epigenetics, Murdoch Children's Research Institute, Royal Children's Hospital, and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Cynthia M de Bont
- Department of Biomolecular Chemistry, Institute for Molecules and Materials (IMM), Radboud University, Nijmegen, the Netherlands
| | - Yutaka Negishi
- Epigenomics & Single Cell Biophysics Group, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ezio T Fok
- Epigenomics & Single Cell Biophysics Group, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Laboratory of Immune Regulation and Tolerance, Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Panayotis Verginis
- Laboratory of Immune Regulation and Tolerance, Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Vera P Mourits
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - L Charlotte J de Bree
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Ger J M Pruijn
- Department of Biomolecular Chemistry, Institute for Molecules and Materials (IMM), Radboud University, Nijmegen, the Netherlands
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans Koenen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Musa M Mhlanga
- Epigenomics & Single Cell Biophysics Group, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dimitri A Diavatopoulos
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands; Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, the Netherlands
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.
| |
Collapse
|
161
|
De Volder J, Vereecke L, Joos G, Maes T. Targeting neutrophils in asthma: A therapeutic opportunity? Biochem Pharmacol 2020; 182:114292. [PMID: 33080186 DOI: 10.1016/j.bcp.2020.114292] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Suppression of airway inflammation with inhaled corticosteroids has been the key therapeutic approach for asthma for many years. Identification of inflammatory phenotypes in asthma has moreover led to important breakthroughs, e.g. with specific targeting of the IL-5 pathway as add-on treatment in difficult-to-treat eosinophilic asthma. However, the impact of interfering with the neutrophilic component in asthma is less documented and understood. This review provides an overview of established and recent insights with regard to the role of neutrophils in asthma, focusing on research in humans. We will describe the main drivers of neutrophilic responses in asthma, the heterogeneity in neutrophils and how they could contribute to asthma pathogenesis. Moreover we will describe findings from clinical trials, in which neutrophilic inflammation was targeted. It is clear that neutrophils are important actors in asthma development and play a role in exacerbations. However, more research is required to fully understand how modulation of neutrophil activity could lead to a significant benefit in asthma patients with airway neutrophilia.
Collapse
Affiliation(s)
- Joyceline De Volder
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lars Vereecke
- VIB Inflammation Research Center, Ghent, Belgium; Ghent Gut Inflammation Group (GGIG), Ghent University, Belgium; Department of Rheumatology, Ghent University Hospital, Belgium
| | - Guy Joos
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Tania Maes
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
162
|
Wang L, Ai Z, Khoyratty T, Zec K, Eames HL, van Grinsven E, Hudak A, Morris S, Ahern D, Monaco C, Eruslanov EB, Luqmani R, Udalova IA. ROS-producing immature neutrophils in giant cell arteritis are linked to vascular pathologies. JCI Insight 2020; 5:139163. [PMID: 32960815 PMCID: PMC7605529 DOI: 10.1172/jci.insight.139163] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022] Open
Abstract
Giant cell arteritis (GCA) is a common form of primary systemic vasculitis in adults, with no reliable indicators of prognosis or treatment responses. We used single cell technologies to comprehensively map immune cell populations in the blood of patients with GCA and identified the CD66b+CD15+CD10lo/–CD64– band neutrophils and CD66bhiCD15+CD10lo/–CD64+/bright myelocytes/metamyelocytes to be unequivocally associated with both the clinical phenotype and response to treatment. Immature neutrophils were resistant to apoptosis, remained in the vasculature for a prolonged period of time, interacted with platelets, and extravasated into the tissue surrounding the temporal arteries of patients with GCA. We discovered that immature neutrophils generated high levels of extracellular reactive oxygen species, leading to enhanced protein oxidation and permeability of endothelial barrier in an in vitro coculture system. The same populations were also detected in other systemic vasculitides. These findings link functions of immature neutrophils to disease pathogenesis, establishing a clinical cellular signature of GCA and suggesting different therapeutic approaches in systemic vascular inflammation. Bona fide immature neutrophil subsets produce unchecked extracellular ROS that contributes to vascular pathologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alison Hudak
- Botnar Research Centre, Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Susan Morris
- Botnar Research Centre, Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Headington, Oxford, United Kingdom
| | | | | | - Evgeniy B Eruslanov
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raashid Luqmani
- Botnar Research Centre, Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Headington, Oxford, United Kingdom
| | | |
Collapse
|
163
|
Liang P, Wang G, Liu X, Wang Z, Wang J, Gao W. Spatiotemporal combination of thermosensitive polypeptide fused interferon and temozolomide for post-surgical glioblastoma immunochemotherapy. Biomaterials 2020; 264:120447. [PMID: 33069137 DOI: 10.1016/j.biomaterials.2020.120447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
Cancer recurrence post surgical resection is of considerable challenge especially in glioblastoma (GBM) therapy. Herein, we demonstrate that interferon-alpha (IFN) fused to a body temperature-sensitive elastin-like polypeptide (IFN-ELP(V)) formed a depot in situ when injected into GBM resection cavity in a mouse brain orthotopic model of GBM. Notably, IFN-ELP(V) in the depot showed a zero-order release kinetics, resulting in dramatically improved pharmacokinetics and biodistribution, and thus inhibited GBM recurrence by stimulating antitumor immunoresponse as compared to IFN. More importantly, when combined with subsequent intraperitoneal injection of temozolomide (TMZ), IFN-ELP(V) could much more effectively suppress post-surgical GBM recurrence than IFN, leading to a remarkably enhanced GBM-free survival rate (60%) over IFN (12.5%). Our findings implicate that the spatiotemporally-programmed combination of IFN-ELP(V) and TMZ leads to the synergy of post-surgical GBM immunochemotherapy, thereby providing a new and effective strategy for cancer therapy.
Collapse
Affiliation(s)
- Ping Liang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Xinyu Liu
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China
| | - Zhuoran Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Jing Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, PR China
| | - Weiping Gao
- Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Biomedical Engineering Department, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
164
|
Habibi MS, Thwaites RS, Chang M, Jozwik A, Paras A, Kirsebom F, Varese A, Owen A, Cuthbertson L, James P, Tunstall T, Nickle D, Hansel TT, Moffatt MF, Johansson C, Chiu C, Openshaw PJM. Neutrophilic inflammation in the respiratory mucosa predisposes to RSV infection. Science 2020; 370:eaba9301. [PMID: 33033192 PMCID: PMC7613218 DOI: 10.1126/science.aba9301] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/30/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
The variable outcome of viral exposure is only partially explained by known factors. We administered respiratory syncytial virus (RSV) to 58 volunteers, of whom 57% became infected. Mucosal neutrophil activation before exposure was highly predictive of symptomatic RSV disease. This was associated with a rapid, presymptomatic decline in mucosal interleukin-17A (IL-17A) and other mediators. Conversely, those who resisted infection showed presymptomatic activation of IL-17- and tumor necrosis factor-related pathways. Vulnerability to infection was not associated with baseline microbiome but was reproduced in mice by preinfection chemokine-driven airway recruitment of neutrophils, which caused enhanced disease mediated by pulmonary CD8+ T cell infiltration. Thus, mucosal neutrophilic inflammation at the time of RSV exposure enhances susceptibility, revealing dynamic, time-dependent local immune responses before symptom onset and explaining the as-yet unpredictable outcomes of pathogen exposure.
Collapse
Affiliation(s)
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Agnieszka Jozwik
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Allan Paras
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Freja Kirsebom
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Augusto Varese
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amber Owen
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Leah Cuthbertson
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Phillip James
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David Nickle
- Genetics & Pharmacogenomics, Department of Translational Medicine, Merck & Co., Inc., Boston, MA, USA
| | - Trevor T Hansel
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| | | |
Collapse
|
165
|
Rubio-Ponce A, Hidalgo A, Ballesteros I. How to bridle a neutrophil. Curr Opin Immunol 2020; 68:41-47. [PMID: 33038850 DOI: 10.1016/j.coi.2020.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022]
Abstract
Recent high-dimensional technologies have enabled the characterization of heterogeneity in the neutrophil compartment at an unprecedented resolution. In this review, we discuss the emerging notion of heterogeneity within the neutrophil pool, and provide a detailed account of evolving concepts in the field. We place special focus on neutrophil differentiation in the bone marrow and plasticity in tissues, describe the limitations that arise when exploring neutrophil heterogeneity using single-cell analyses, and suggest state-of-the-art alternatives to improve their characterization. Finally, we propose strategies arising from these new concepts that may allow us to bridle neutrophil plasticity towards therapeutic benefit.
Collapse
Affiliation(s)
- Andrea Rubio-Ponce
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Andrés Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Iván Ballesteros
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.
| |
Collapse
|
166
|
Sun X, Ni N, Ma Y, Wang Y, Leong DT. Retooling Cancer Nanotherapeutics' Entry into Tumors to Alleviate Tumoral Hypoxia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003000. [PMID: 32803846 DOI: 10.1002/smll.202003000] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/20/2020] [Indexed: 06/11/2023]
Abstract
Anti-hypoxia cancer nanomedicine (AHCN) holds exciting potential in improving oxygen-dependent therapeutic efficiencies of malignant tumors. However, most studies regarding AHCN focus on optimizing structure and function of nanomaterials with presupposed successful entry into tumor cells. From such a traditional perspective, the main barrier that AHCN needs to overcome is mainly the tumor cell membrane. However, such an oversimplified perspective would neglect that real tumors have many biological, physiological, physical, and chemical defenses preventing the current state-of-the-art AHCNs from even reaching the targeted tumor cells. Fortunately, in recent years, some studies are beginning to intentionally focus on overcoming physiological barriers to alleviate hypoxia. In this Review, the limitations behind the traditional AHCN delivery mindset are addressed and the key barriers that need to be surmounted before delivery to cancer cells and some good ways to improve cell membrane attachment, internalization, and intracellular retention are summarized. It is aimed to contribute to Review literature on this emerging topic through refreshing perspectives based on this work and what is also learnt from others. This Review would therefore assist AHCNs researchers to have a quick overview of the essential information and glean thought-provoking ideas to advance this sub-field in cancer nanomedicine.
Collapse
Affiliation(s)
- Xiao Sun
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yanling Ma
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yan Wang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
167
|
Xu L, Jin Y, Qin X. Comprehensive analysis of significant genes and immune cell infiltration in HPV-related head and neck squamous cell carcinoma. Int Immunopharmacol 2020; 87:106844. [DOI: 10.1016/j.intimp.2020.106844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/28/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
|
168
|
van Elsas M, Kleinovink JW, Moerland M, Feiss G, Beyrend G, Arens R, Mei H, Nibbering PH, Jirka SM, van Hall T, van der Burg SH. Host genetics and tumor environment determine the functional impact of neutrophils in mouse tumor models. J Immunother Cancer 2020; 8:jitc-2020-000877. [PMID: 32998952 PMCID: PMC7528431 DOI: 10.1136/jitc-2020-000877] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2020] [Indexed: 01/06/2023] Open
Abstract
Background Neutrophils have been reported to have protumor, antitumor or neutral effects in cancer progression. The underlying causes for this functional variability are not clear. Methods We studied the role of neutrophils in six different mouse tumor models by intratumoral injection of antimicrobial peptides or vaccination. Changes in systemic and intratumoral immune cells were analyzed by flow-cytometry and mass-cytometry. The role of neutrophils was studied by antibody-mediated neutrophil depletion. Neutrophils from different mouse strains were compared by RNA sequencing. Results The antimicrobial peptide Omiganan reduced the growth of TC-1 tumors in BL/6 mice and CT26 tumors in BALB/c mice. No significant effects were observed in B16F10, MC38 and 4T1 tumors. Growth delay was associated with increased abundance of neutrophils in TC-1 but not CT26 tumors. Systemic neutrophil depletion abrogated Omiganan efficacy in TC-1 but further reduced growth of CT26, indicating that neutrophils were required for the antitumor effect in TC-1 but suppressed tumor control in CT26. Neutrophils were also required for a therapeutic vaccine-induced T-cell mediated control of RMA tumors in BL/6 mice. Clearly, the circulating and intratumoral neutrophils differed in the expression of Ly6G and CD62L, between TC-1 and CT26 and between blood neutrophils of tumor-naïve BL/6 and BALB/c mice. RNA-sequencing revealed that neutrophils from BL/6 mice but not BALB/c mice displayed a robust profile of immune activation, matching their opposing roles in TC-1 and RMA versus CT26. Conclusions Neutrophil functionality differs strongly between mouse strains and tumor types, with consequences for tumor progression and therapy.
Collapse
Affiliation(s)
- Marit van Elsas
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Willem Kleinovink
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Gary Feiss
- Cutanea Life Sciences, Wayne, Pennsylvania, USA
| | - Guillaume Beyrend
- Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Arens
- Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter H Nibbering
- Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Thorbald van Hall
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
169
|
Kappes L, Amer RL, Sommerlatte S, Bashir G, Plattfaut C, Gieseler F, Gemoll T, Busch H, Altahrawi A, Al-Sbiei A, Haneefa SM, Arafat K, Schimke LF, Khawanky NE, Schulze-Forster K, Heidecke H, Kerstein-Staehle A, Marschner G, Pitann S, Ochs HD, Mueller A, Attoub S, Fernandez-Cabezudo MJ, Riemekasten G, Al-Ramadi BK, Cabral-Marques O. Ambrisentan, an endothelin receptor type A-selective antagonist, inhibits cancer cell migration, invasion, and metastasis. Sci Rep 2020; 10:15931. [PMID: 32985601 PMCID: PMC7522204 DOI: 10.1038/s41598-020-72960-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/07/2020] [Indexed: 12/17/2022] Open
Abstract
Several studies reported a central role of the endothelin type A receptor (ETAR) in tumor progression leading to the formation of metastasis. Here, we investigated the in vitro and in vivo anti-tumor effects of the FDA-approved ETAR antagonist, Ambrisentan, which is currently used to treat patients with pulmonary arterial hypertension. In vitro, Ambrisentan inhibited both spontaneous and induced migration/invasion capacity of different tumor cells (COLO-357 metastatic pancreatic adenocarcinoma, OvCar3 ovarian carcinoma, MDA-MB-231 breast adenocarcinoma, and HL-60 promyelocytic leukemia). Whole transcriptome analysis using RNAseq indicated Ambrisentan's inhibitory effects on the whole transcriptome of resting and PAR2-activated COLO-357 cells, which tended to normalize to an unstimulated profile. Finally, in a pre-clinical murine model of metastatic breast cancer, treatment with Ambrisentan was effective in decreasing metastasis into the lungs and liver. Importantly, this was associated with a significant enhancement in animal survival. Taken together, our work suggests a new therapeutic application for Ambrisentan in the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Lucy Kappes
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Ruba L Amer
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sabine Sommerlatte
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Ghada Bashir
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Corinna Plattfaut
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Lübeck, Lübeck, Germany
| | - Frank Gieseler
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Lübeck, Lübeck, Germany
| | - Timo Gemoll
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hauke Busch
- Lübeck Institute for Experimental Dermatology (LIED) and Institute of Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Abeer Altahrawi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ashraf Al-Sbiei
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Shoja M Haneefa
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kholoud Arafat
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Lena F Schimke
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Nadia El Khawanky
- Department of Hematology and Oncology, Faculty of Medicine, The University of Freiburg, Freiburg, Germany
| | - Kai Schulze-Forster
- CellTrend GmbH, Luckenwalde, Brandenburg, Germany
- Department of Urology, Charité University Hospital, Berlin, Germany
| | | | - Anja Kerstein-Staehle
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Gabriele Marschner
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Silke Pitann
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, USA
| | - Antje Mueller
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Samir Attoub
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Maria J Fernandez-Cabezudo
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Lineu Prestes Avenue, 1730, São Paulo, SP, Brazil.
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, Brazil.
| |
Collapse
|
170
|
Wang Z, Yang C, Li L, Jin X, Zhang Z, Zheng H, Pan J, Shi L, Jiang Z, Su K, Li B, Shao X, Qiu F, Yan J, Huang J. Tumor-derived HMGB1 induces CD62L dim neutrophil polarization and promotes lung metastasis in triple-negative breast cancer. Oncogenesis 2020; 9:82. [PMID: 32943604 PMCID: PMC7499196 DOI: 10.1038/s41389-020-00267-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/23/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive, difficult to treat and commonly develops visceral metastasis, including lung metastasis. We observed that High mobility group box 1 protein (HMGB1) was highly expressed in human TNBC and positively correlated with cancer metastasis. The hypoxic tumor environment is known to regulate HMGB1 secretion, but an understanding of the underlying mechanism by which tumor-derived HMGB1 regulates interstitial components and promotes breast cancer lung metastasis has remained elusive. The results of the present study showed that the number of CD62Ldim neutrophils, which have a strong ability to produce neutrophil extracellular traps (NETs), increased significantly in both peripheral blood and lung tissues in a mouse TNBC model and were regulated by tumor-derived HMGB1 through the TLR2 pathway. Furthermore, serum HMGB1 levels were positively correlated with CD62Ldim neutrophils in 86 breast cancer patients. We demonstrated that CD62Ldim neutrophils accelerated lung metastasis and that interventions targeting the “HMGB1-CD62Ldim neutrophil-NETs” axis could inhibit lung metastasis. Our results suggest that the combination of HMGB1 and CD62Ldim neutrophils is a potential marker for breast cancer lung metastasis and is novel target for future prevention and therapy.
Collapse
Affiliation(s)
- Zhen Wang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chenghui Yang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lili Li
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiaoyan Jin
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Surgical Oncology, Zhejiang Taizhou Municipal Hospital, Taizhou, 318008, China
| | - Zhigang Zhang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Gynecology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haiyan Zheng
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jun Pan
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Liyun Shi
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhou Jiang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ke Su
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Baizhou Li
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xuan Shao
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fuming Qiu
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jun Yan
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Jian Huang
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
171
|
Vergnano M, Mockenhaupt M, Benzian-Olsson N, Paulmann M, Grys K, Mahil SK, Chaloner C, Barbosa IA, August S, Burden AD, Choon SE, Cooper H, Navarini AA, Reynolds NJ, Wahie S, Warren RB, Wright A, Huffmeier U, Baum P, Visvanathan S, Barker JN, Smith CH, Capon F. Loss-of-Function Myeloperoxidase Mutations Are Associated with Increased Neutrophil Counts and Pustular Skin Disease. Am J Hum Genet 2020; 107:539-543. [PMID: 32758448 PMCID: PMC7477255 DOI: 10.1016/j.ajhg.2020.06.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
The identification of disease alleles underlying human autoinflammatory diseases can provide important insights into the mechanisms that maintain neutrophil homeostasis. Here, we focused our attention on generalized pustular psoriasis (GPP), a potentially life-threatening disorder presenting with cutaneous and systemic neutrophilia. Following the whole-exome sequencing of 19 unrelated affected individuals, we identified a subject harboring a homozygous splice-site mutation (c.2031-2A>C) in MPO. This encodes myeloperoxidase, an essential component of neutrophil azurophil granules. MPO screening in conditions phenotypically related to GPP uncovered further disease alleles in one subject with acral pustular psoriasis (c.2031-2A>C;c.2031-2A>C) and in two individuals with acute generalized exanthematous pustulosis (c.1705C>T;c.2031-2A>C and c.1552_1565del;c.1552_1565del). A subsequent analysis of UK Biobank data demonstrated that the c.2031-2A>C and c.1705C>T (p.Arg569Trp) disease alleles were also associated with increased neutrophil abundance in the general population (p = 5.1 × 10-6 and p = 3.6 × 10-5, respectively). The same applied to three further deleterious variants that had been genotyped in the cohort, with two alleles (c.995C>T [p.Ala332Val] and c.752T>C [p.Met251Thr]) yielding p values < 10-10. Finally, treatment of healthy neutrophils with an MPO inhibitor (4-Aminobenzoic acid hydrazide) increased cell viability and delayed apoptosis, highlighting a mechanism whereby MPO mutations affect granulocyte numbers. These findings identify MPO as a genetic determinant of pustular skin disease and neutrophil abundance. Given the recent interest in the development of MPO antagonists for the treatment of neurodegenerative disease, our results also suggest that the pro-inflammatory effects of these agents should be closely monitored.
Collapse
Affiliation(s)
- Marta Vergnano
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK; St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Maja Mockenhaupt
- Department of Dermatology, Medical Centre-University of Freiburg, Freiburg 79106, Germany
| | - Natashia Benzian-Olsson
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Maren Paulmann
- Department of Dermatology, Medical Centre-University of Freiburg, Freiburg 79106, Germany
| | - Katarzyna Grys
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Satveer K Mahil
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Charlotte Chaloner
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Ines A Barbosa
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | | | - A David Burden
- Department of Dermatology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Siew-Eng Choon
- Department of Dermatology, Sultanah Aminah Hospital, Clinical School Johor Bahru, Monash University, Malaysia
| | - Hywel Cooper
- Portsmouth Dermatology Centre, St Marys Hospital, Portsmouth PO3 6AD, UK
| | - Alex A Navarini
- Department of Dermatology & Allergy, University Hospital of Basel, Basel 4031, Switzerland
| | - Nick J Reynolds
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK and Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
| | - Shyamal Wahie
- Department of Dermatology, University Hospital of North Durham, Durham DH1 5TW, UK
| | - Richard B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester NIHR Biomedical Research Centre, University of Manchester, Manchester M6 8HD, UK
| | - Andrew Wright
- Centre for Skin Sciences, St Lukes Hospital, Bradford BD5 0NA, UK
| | - Ulrike Huffmeier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Patrick Baum
- Boehringer-Ingelheim International GmbH, Biberach 88397, Germany
| | | | - Jonathan N Barker
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Catherine H Smith
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK
| | - Francesca Capon
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London SE1 9RT, UK.
| |
Collapse
|
172
|
Tumour-associated neutrophils orchestrate intratumoural IL-8-driven immune evasion through Jagged2 activation in ovarian cancer. Br J Cancer 2020; 123:1404-1416. [PMID: 32778818 PMCID: PMC7591527 DOI: 10.1038/s41416-020-1026-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 07/07/2020] [Accepted: 07/23/2020] [Indexed: 01/22/2023] Open
Abstract
Background Tumour associated neutrophils (TANs) play a controversial role in regulating immune surveillance and immune evasion in various malignancies. Here, we investigated the relevance of TANs with the prognosis and immune microenvironment of epithelial ovarian cancer (EOC). Methods We characterised TANs using flow cytometric analysis and immunofluorescence analysis. The prognostic merit of TANs in EOC was evaluated using cox regression analysis. Furthermore, we explored the therapeutic merit of targeting Notch signalling in EOC and determined its involvement in the immune microenvironment. Results High level of TANs is associated with a dismal prognosis and immune tolerance in EOC. TANs impaired cytotoxic effects of CD8+ T cells partly through Jagged2 (JAG2). Notch pathway blocked using γ-secretase inhibitor LY3039478 and anti-JAG2 antibody led to retarded tumour growth and augmented cytotoxic effects of CD8+ T cells. IL-8 contributes to the recruitment of TANs and the induction of JAG2 expression in TANs. Blockade of CXCR2 signalling reduces tumour growth rate, accompanied by a decreasing amount of TANs and increasing activity of CD8+ T cells. JAG2+TANs is an independent predictor of clinical outcomes. Conclusion JAG2+TANs are closely linked to IL-8-driven immune evasion microenvironment and may serve as a promising therapeutic target for the reinvigoration of anti-tumour immunity.
Collapse
|
173
|
Zhang W, Shen Y, Huang H, Pan S, Jiang J, Chen W, Zhang T, Zhang C, Ni C. A Rosetta Stone for Breast Cancer: Prognostic Value and Dynamic Regulation of Neutrophil in Tumor Microenvironment. Front Immunol 2020; 11:1779. [PMID: 32849640 PMCID: PMC7426521 DOI: 10.3389/fimmu.2020.01779] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence has revealed that the initiation and progression of breast cancer are greatly affected by the immune environment. Neutrophils are the most abundant leucocytes in circulation and act as the spearhead in inflammation, including in breast cancer. Circulating neutrophils are closely related to the prognosis of breast cancer patients, and tumor-infiltrating neutrophils have varied functions at different stages of breast cancer, such as antitumor or tumor-promoting neutrophils, which are termed N1 and N2 neutrophils, respectively. In this review, we will discuss the utility of circulating neutrophils for predicting prognosis and therapeutic efficacy and the underlying mechanisms of their chemotaxis, the dynamic regulation of their antitumor or protumor functions and their different spatial distributions in tumor microenvironment. Finally, we also discuss the possibility of targeting neutrophils as a therapeutic strategy in breast cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Endocrinology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yimin Shen
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Huanhuan Huang
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Sheng Pan
- School of Medicine, Chu Kochen Honors College, Zhejiang University, Hangzhou, China
| | - Jingxin Jiang
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Wuzhen Chen
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ting Zhang
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Chao Zhang
- Department of Anatomy, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chao Ni
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
174
|
Tang L, Wang Z, Mu Q, Yu Z, Jacobson O, Li L, Yang W, Huang C, Kang F, Fan W, Ma Y, Wang M, Zhou Z, Chen X. Targeting Neutrophils for Enhanced Cancer Theranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002739. [PMID: 32656801 DOI: 10.1002/adma.202002739] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/18/2020] [Indexed: 05/18/2023]
Abstract
Improving tumor accumulation and delivery efficiency is an important goal of nanomedicine. Neutrophils play a vital role in both chemically mediating inflammatory response through myeloperoxidase (MPO) and biologically promoting metastasis during inflammation triggered by the primary tumor or environmental stimuli. Herein, a novel theranostic nanomedicine that targets both the chemical and biological functions of neutrophils in tumor is designed, facilitating the enhanced retention and sustained release of drug cargos for improved cancer theranostics. 5-hydroxytryptamine (5-HT) is equipped onto nanoparticles (NPs) loaded with photosensitizers and Zileuton (a leukotriene inhibitor) to obtain MPO and neutrophil targeting NPs, denoted as HZ-5 NPs. The MPO targeting property of 5-HT modified NPs is confirmed by noninvasive positron emission tomography imaging studies. Furthermore, photodynamic therapy is used to initiate the inflammatory response which further mediated the accumulation and retention of neutrophil targeting NPs in a breast cancer model. This design renders a greatly improved theranostic nanomedicine for efficient tumor suppression, and more importantly, inhibition of neutrophil-mediated lung metastasis via the sustained release of Zileuton. This work presents a novel strategy of targeting neutrophils for improved tumor theranostics, which may open up new avenues in designing nanomedicine through exploiting the tumor microenvironment.
Collapse
Affiliation(s)
- Longguang Tang
- The People's Hospital of Gaozhou, Maoming, 525200, China
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qingchun Mu
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ling Li
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weijing Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chunming Huang
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Fei Kang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ying Ma
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maosheng Wang
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
175
|
Theodros D, Murter BM, Sidhom JW, Nirschl TR, Clark DJ, Chen L, Tam AJ, Blosser RL, Schwen ZR, Johnson MH, Pierorazio PM, Zhang H, Ganguly S, Pardoll DM, Zarif JC. High-dimensional Cytometry (ExCYT) and Mass Spectrometry of Myeloid Infiltrate in Clinically Localized Clear Cell Renal Cell Carcinoma Identifies Novel Potential Myeloid Targets for Immunotherapy. Mol Cell Proteomics 2020; 19:1850-1859. [PMID: 32737216 PMCID: PMC7664124 DOI: 10.1074/mcp.ra120.002049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/24/2020] [Indexed: 01/05/2023] Open
Abstract
Although the focus of the role of cancer immunotherapy has been in advanced disease states, we sought to investigate changes to the immune infiltrate of early, clinically localized clear cell Renal Cell Carcinoma (ccRCC). Using orthogonal approaches including Mass Spectrometry on immune cell infiltrates, we report numerous alterations that provide new insight into the biology of treatment-naïve ccRCC and identification of novel targets that may prove to be clinically impactful. Renal Cell Carcinoma (RCC) is one of the most commonly diagnosed cancers worldwide with research efforts dramatically improving understanding of the biology of the disease. To investigate the role of the immune system in treatment-naïve clear cell Renal Cell Carcinoma (ccRCC), we interrogated the immune infiltrate in patient-matched ccRCC tumor samples, benign normal adjacent tissue (NAT) and peripheral blood mononuclear cells (PBMCs isolated from whole blood, focusing our attention on the myeloid cell infiltrate. Using flow cytometric, MS, and ExCYT analysis, we discovered unique myeloid populations in PBMCs across patient samples. Furthermore, normal adjacent tissues and ccRCC tissues contained numerous myeloid populations with a unique signature for both tissues. Enrichment of the immune cell (CD45+) fraction and subsequent gene expression analysis revealed a number of myeloid-related genes that were differentially expressed. These data provide evidence, for the first time, of an immunosuppressive and pro-tumorigenic role of myeloid cells in early, clinically localized ccRCC. The identification of a number of immune proteins for therapeutic targeting provides a rationale for investigation into the potential efficacy of earlier intervention with single-agent or combination immunotherapy for ccRCC.
Collapse
Affiliation(s)
- Debebe Theodros
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benjamin M Murter
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John-William Sidhom
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas R Nirschl
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Pathobiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - David J Clark
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - LiJun Chen
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ada J Tam
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Richard L Blosser
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Zeyad R Schwen
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael H Johnson
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Phillip M Pierorazio
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sudipto Ganguly
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Drew M Pardoll
- Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Pathobiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Jelani C Zarif
- Bloomberg-Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA.
| |
Collapse
|
176
|
Alipour R, Motedayyen H, Sereshki N, Rafiee M, Alsahebfosul F, Pourazar A. Human Amniotic Epithelial Cells Affect the Functions of Neutrophils. Int J Stem Cells 2020; 13:212-220. [PMID: 32323513 PMCID: PMC7378904 DOI: 10.15283/ijsc19155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Objectives As a stem cell group, Human amniotic epithelial cells (HAECs) have numerous advantages over their embryonic and adult counterparts for therapeutic utility. They are closer to clinical applications compared to other stem cell types. Additionally, the anti-inflammatory and immunoregulatory properties of HAECs toward several immune cells have been shown previously. Nevertheless, despite the ever-increasing importance of neutrophils in the immune and non-immune processes, a few studies investigated the interaction of neutrophils and HAECs. To increase the current knowledge of HAECs immunology which is necessary for optimizing their future clinical applications, here we explored the effect of HAECs on two chief neutrophil functions; respiratory burst and phagocytosis. Methods and Results Freshly isolated human blood neutrophils were co-cultured with different number of HAECs for about 24 or 48 hours, then the oxidative burst and phagocytosis of stimulated neutrophils were assessed and compared. The results demonstrated a substantial elevation in the phagocytosis percentage, conversely a significant reduction in the oxidative burst of HAECs-cocultured neutrophils. These effects were dose-dependent, but did not show similar patterns. Likewise, the elongation of coculture period inversely influenced the HAECs-induced effects on the two neutrophil functions. Conclusions The present study, for the first time, investigated the HAECs-mediated effects on the two main neutrophil functions. The findings suggest that HAECs by enhancement of phagocytic ability and simultaneously, attenuation of oxidative burst capacity of neutrophils protect the fetus from both microbial treats and oxidative stress and their consequent inflammation; thus corroborate the current anti-inflammatory vision of HAECs.
Collapse
Affiliation(s)
- Razieh Alipour
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Motedayyen
- Department of Immunology, Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Nasrin Sereshki
- Department of Immunology, Asadabad School of Medical Science, Asadabad, Iran
| | - Mitra Rafiee
- Cellular and Molecular Research Center, Department of Immunology, Medical School, Birjand University of Medical Sciences, Birjand, Iran
| | - Fereshteh Alsahebfosul
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbasali Pourazar
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
177
|
Xie X, Shi Q, Wu P, Zhang X, Kambara H, Su J, Yu H, Park SY, Guo R, Ren Q, Zhang S, Xu Y, Silberstein LE, Cheng T, Ma F, Li C, Luo HR. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol 2020; 21:1119-1133. [PMID: 32719519 PMCID: PMC7442692 DOI: 10.1038/s41590-020-0736-z] [Citation(s) in RCA: 375] [Impact Index Per Article: 93.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 06/11/2020] [Indexed: 01/10/2023]
Abstract
The full neutrophil heterogeneity and differentiation landscape remains incompletely characterized. Here we profiled >25,000 differentiating and mature mouse neutrophils using single-cell RNA sequencing to provide a comprehensive transcriptional landscape of neutrophil maturation, function, and fate decision in their steady state and during bacterial infection. Eight neutrophil populations were defined by distinct molecular signatures. The three mature peripheral blood neutrophil subsets arise from distinct maturing bone marrow neutrophil subsets. Driven by both known and uncharacterized transcription factors, neutrophils gradually acquire microbicidal capability as they traverse the transcriptional landscape, representing an evolved mechanism for fine-tuned regulation of an effective but balanced neutrophil response. Bacterial infection reprograms the genetic architecture of neutrophil populations, alters dynamic transition between each subpopulation, and primes neutrophils for augmented functionality without affecting overall heterogeneity. In summary, these data establish a reference model and general framework for studying neutrophil-related disease mechanisms, biomarkers, and therapeutic targets at single-cell resolution.
Collapse
Affiliation(s)
- Xuemei Xie
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Qiang Shi
- School of Life Sciences, Center for Bioinformatics, Center for Statistical Science, Peking University, Beijing, China
| | - Peng Wu
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaoyu Zhang
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Hiroto Kambara
- Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Jiayu Su
- Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Dana-Farber/Harvard Cancer Center, Boston, MA, USA.,School of Life Sciences, Center for Bioinformatics, Center for Statistical Science, Peking University, Beijing, China
| | - Hongbo Yu
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shin-Young Park
- Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Rongxia Guo
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qian Ren
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Sudong Zhang
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuanfu Xu
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Leslie E Silberstein
- Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.,Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Tao Cheng
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fengxia Ma
- The State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Cheng Li
- School of Life Sciences, Center for Bioinformatics, Center for Statistical Science, Peking University, Beijing, China.
| | - Hongbo R Luo
- Department of Pathology, Harvard Medical School, Boston, MA, USA. .,Department of Laboratory Medicine, The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA. .,Dana-Farber/Harvard Cancer Center, Boston, MA, USA.
| |
Collapse
|
178
|
Lim CL, Or YZ, Ong Z, Chung HH, Hayashi H, Shrestha S, Chiba S, Lin F, Lin VCL. Estrogen exacerbates mammary involution through neutrophil-dependent and -independent mechanism. eLife 2020; 9:57274. [PMID: 32706336 PMCID: PMC7417171 DOI: 10.7554/elife.57274] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
There is strong evidence that the pro-inflammatory microenvironment during post-partum mammary involution promotes parity-associated breast cancer. Estrogen exposure during mammary involution drives tumor growth through neutrophils’ activity. However, how estrogen and neutrophils influence mammary involution are unknown. Combined analysis of transcriptomic, protein, and immunohistochemical data in BALB/c mice showed that estrogen promotes involution by exacerbating inflammation, cell death and adipocytes repopulation. Remarkably, 88% of estrogen-regulated genes in mammary tissue were mediated through neutrophils, which were recruited through estrogen-induced CXCR2 signalling in an autocrine fashion. While neutrophils mediate estrogen-induced inflammation and adipocytes repopulation, estrogen-induced mammary cell death was via lysosome-mediated programmed cell death through upregulation of cathepsin B, Tnf and Bid in a neutrophil-independent manner. Notably, these multifaceted effects of estrogen are mostly mediated by ERα and unique to the phase of mammary involution. These findings are important for the development of intervention strategies for parity-associated breast cancer.
Collapse
Affiliation(s)
- Chew Leng Lim
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yu Zuan Or
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Zoe Ong
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Hwa Hwa Chung
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Hirohito Hayashi
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, Singapore
| | - Smeeta Shrestha
- School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore, India
| | - Shunsuke Chiba
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, Singapore
| | - Feng Lin
- School of Computer Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Valerie Chun Ling Lin
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
179
|
Mroweh M, Decaens T, Marche PN, Macek Jilkova Z, Clément F. Modulating the Crosstalk between the Tumor and Its Microenvironment Using RNA Interference: A Treatment Strategy for Hepatocellular Carcinoma. Int J Mol Sci 2020; 21:E5250. [PMID: 32722054 PMCID: PMC7432232 DOI: 10.3390/ijms21155250] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy with one of the highest mortality rates among solid cancers. It develops almost exclusively in the background of chronic liver inflammation, which can be caused by viral hepatitis, chronic alcohol consumption or an unhealthy diet. Chronic inflammation deregulates the innate and adaptive immune responses that contribute to the proliferation, survival and migration of tumor cells. The continuous communication between the tumor and its microenvironment components serves as the overriding force of the tumor against the body's defenses. The importance of this crosstalk between the tumor microenvironment and immune cells in the process of hepatocarcinogenesis has been shown, and therapeutic strategies modulating this communication have improved the outcomes of patients with liver cancer. To target this communication, an RNA interference (RNAi)-based approach can be used, an innovative and promising strategy that can disrupt the crosstalk at the transcriptomic level. Moreover, RNAi offers the advantage of specificity in comparison to the treatments currently used for HCC in clinics. In this review, we will provide the recent data pertaining to the modulation of a tumor and its microenvironment by using RNAi and its potential for therapeutic intervention in HCC.
Collapse
Affiliation(s)
- Mariam Mroweh
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (T.D.); (P.N.M.)
- Université Grenoble Alpes, 38000 Grenoble, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath Beirut 6573-14, Lebanon
| | - Thomas Decaens
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (T.D.); (P.N.M.)
- Université Grenoble Alpes, 38000 Grenoble, France
- Service d’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Patrice N Marche
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (T.D.); (P.N.M.)
- Université Grenoble Alpes, 38000 Grenoble, France
| | - Zuzana Macek Jilkova
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (T.D.); (P.N.M.)
- Université Grenoble Alpes, 38000 Grenoble, France
- Service d’hépato-Gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Flora Clément
- Institute for Advanced Biosciences, Research Center Inserm U 1209/CNRS 5309, 38700 La Tronche, France; (M.M.); (T.D.); (P.N.M.)
- Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
180
|
Zucker A, Winter A, Lumley D, Karwowski P, Jung MK, Kao J. Prognostic role of baseline neutrophil-to-lymphocyte ratio in metastatic solid tumors. Mol Clin Oncol 2020; 13:25. [PMID: 32774855 DOI: 10.3892/mco.2020.2095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/15/2020] [Indexed: 01/04/2023] Open
Abstract
High baseline neutrophil-to-lymphocyte ratio (NLR) has been associated with poor survival in a number of solid tumors, but has not been extensively investigated in the context of radiation oncology. Developing more robust models to predict survival would inform patient care for patients with metastatic solid tumors. The present study was undertaken to evaluate the effect of baseline NLR (using 4 as a cutoff) on survival in 320 consecutive patients with metastatic cancer who were referred to a single radiation oncologist between 2012 and 2015, with a median follow-up of 20.6 months. The median NLR was 4.4 (interquartile range, 2.8-7.2). Patients with a baseline NLR ≤4 had a median survival of 9.3 months compared to 4.1 months for NLR >4 (P<0.001). The number of active tumors, Eastern Cooperative Oncology Group performance status score, baseline albumin, primary tumor site, liver metastases and baseline NLR predicted overall survival on both univariate and multivariate analysis (P<0.05 for all). After adjusting for known prognostic factors for advanced solid tumors, baseline NLR >4 independently predicted adverse survival in this cohort.
Collapse
Affiliation(s)
- Amanda Zucker
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Alex Winter
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Dean Lumley
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Pawel Karwowski
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Min-Kyung Jung
- College of Osteopathic Medicine, New York Institute of Technology, Glen Head, NY 11545, USA
| | - Johnny Kao
- Department of Radiation Oncology, Good Samaritan Hospital Medical Center, West Islip, NY 11795, USA
| |
Collapse
|
181
|
Roles of the Site 2 Protease Eep in Staphylococcus aureus. J Bacteriol 2020; 202:JB.00046-20. [PMID: 32457050 DOI: 10.1128/jb.00046-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/21/2020] [Indexed: 01/02/2023] Open
Abstract
In Enterococcus faecalis, the site 2 protease Eep generates sex pheromones, including cAM373. Intriguingly, in Staphylococcus aureus, a peptide similar to cAM373, named cAM373_SA, is produced from the camS gene. Here, we report that the staphylococcal Eep homolog is not only responsible for the production of cAM373_SA but also critical for staphylococcal virulence. As with other Eep proteins, the staphylococcal Eep protein has four transmembrane (TM) domains, with the predicted zinc metalloprotease active site (HEXXH) in the first TM domain. eep deletion reduced the cAM373_SA activity in the culture supernatant to the level of the camS deletion mutant. It also markedly decreased the cAM373 peptide peak in a high-performance liquid chromatography (HPLC) analysis. Proteomics analysis showed that Eep affects the production and/or the release of diverse proteins, including the signal peptidase subunit SpsB and the surface proteins SpA, SasG, and FnbA. eep deletion decreased the adherence of S. aureus to host epithelial cells; however, the adherence of the eep mutant was increased by overexpression of the surface proteins SpA, SasG, and FnbA. eep deletion reduced staphylococcal resistance to killing by human neutrophils as well as survival in a murine model of blood infection. The overexpression of the surface protein SpA in the eep mutant increased bacterial survival in the liver. Our study illustrates that in S. aureus, Eep not only generates cAM373_SA but also contributes to the survival of the bacterial pathogen in the host.IMPORTANCE The emergence of multidrug-resistant Staphylococcus aureus makes the treatment of staphylococcal infections much more difficult. S. aureus can acquire a drug resistance gene from other bacteria, such as Enterococcus faecalis Intriguingly, S. aureus produces a sex pheromone for the E. faecalis plasmid pAM373, raising the possibility that S. aureus actively promotes plasmid conjugation from E. faecalis In this study, we found that the staphylococcal Eep protein is responsible for sex pheromone processing and contributes to the survival of the bacteria in the host. These results will enhance future research on the drug resistance acquisition of S. aureus and can lead to the development of novel antivirulence drugs.
Collapse
|
182
|
Capucetti A, Albano F, Bonecchi R. Multiple Roles for Chemokines in Neutrophil Biology. Front Immunol 2020; 11:1259. [PMID: 32733442 PMCID: PMC7363767 DOI: 10.3389/fimmu.2020.01259] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 11/23/2022] Open
Abstract
Chemokines are recognized as the most critical mediators for selective neutrophil recruitment during inflammatory conditions. Furthermore, they are considered fundamental regulators of neutrophil mobilization from the bone marrow (BM) to the bloodstream and for their homing back at the end of their life for apoptosis and clearance. However, chemokines are also important mediators of neutrophil effector functions including oxidative burst, degranulation, neutrophil extracellular trap (NET)osis, and production of inflammatory mediators. Neutrophils have been historically considered as a homogeneous population. In recent years, several maturation stages and subsets with different phenotypic profiles and effector functions were described both in physiological and pathological conditions such as infections, autoimmunity, and cancer. The aim of this review is to give an overview of the current evidence regarding the role of chemokines and chemokine receptors in neutrophil biology, including their possible role in neutrophil maturation, differentiation, and in defining emerging neutrophil subsets.
Collapse
Affiliation(s)
- Arianna Capucetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Francesca Albano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| |
Collapse
|
183
|
Makatsariya AD, Slukhanchuk EV, Bitsadze VO, Khizroeva JKH, Tretyakova MV, Tsibizova VI, Elalamy I, Gris JC, Grandone E, Makatsariya NA, Mashkova T. Thrombotic microangiopathy, DIC-syndrome and COVID-19: link with pregnancy prothrombotic state. J Matern Fetal Neonatal Med 2020; 35:2536-2544. [PMID: 32627622 DOI: 10.1080/14767058.2020.1786811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
For last months, humanity has faced a formidable unknown enemy, which is presented as a new coronavirus infection. Despite the fact that the causative agents of new diseases appear at a certain frequency and that the virus SARS-CoV-2 has certain common properties with its predecessors, at the moment we are dealing with a new unknown pathogenesis of the development of severe complications in patients with risk factors. A final understanding of pathological process mechanisms is the goal of the scientific community. Summarizing research data from different countries, it became obvious that in severe cases of viral infection, we are dealing with a combination of the systemic inflammatory response syndrome, disseminated intravascular coagulation and thrombotic microangiopathy (TMA). Thrombotic microangiopathy is represented by a group of different conditions in which thrombocytopenia, hemolytic anemia, and multiple organ failure occur. The article reflects the main types of TMA, pathogenesis and principles of therapy. The main participants in the process are described in detail, including the von Willebrand factor and ADAMTS-13. Based on the knowledge available, as well as new data obtained from patients with COVID-19, we proposed possible models for the implementation of conditions such as sepsis, TMA, and DIC in patients with severe new coronavirus infection. Through a deeper understanding of pathogenesis, it will be possible to develop more effective diagnosis and therapy.
Collapse
Affiliation(s)
- A D Makatsariya
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia
| | - E V Slukhanchuk
- Head of Gynecology Unit, Petrovsky National Research Center of Surgery, Moscow, Russia
| | - V O Bitsadze
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia
| | - J K H Khizroeva
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia
| | - M V Tretyakova
- Gynecology Department, «Medical Center» LLC, Moscow, Russia
| | - V I Tsibizova
- Almazov National Medical Research Centre, Saint Petersburg, Health Ministry of Russian Federation, Saint Petersburg, Russia
| | - I Elalamy
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia.,Director of Hematology Department of Thrombosis Center, Medicine Sorbonne University, Paris, France
| | - J-C Gris
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia.,University Montpellier, Montpellier, France
| | - E Grandone
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia.,Hemostasis and Thrombosis Unit, Fondazione I.R.C.C.S. "Casa Sollievo della Sofferenza", Puglia, Italy
| | - N A Makatsariya
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia
| | - T Mashkova
- Obstetrics and Gynecology, The First I.M. Sechenov Moscow State Medical University, (Sechenov University), Moscow, Russia
| |
Collapse
|
184
|
Li Y, Wang D, Li X. The blood cells in NSCLC and the changes after RFA. Int J Hyperthermia 2020; 37:753-762. [PMID: 32619369 DOI: 10.1080/02656736.2020.1782486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Lung cancer has attracted a lot of attention because of its high morbidity and mortality. The emergence of RFA provides a new treatment for unresectable NSCLC patients. In addition to killing in situ lung tumors, RFA also provides new immuno-activated antigens, for the treatment of lung cancer. It changes the tumor microenvironment and activates the entire immune system of patients. The peripheral blood cell count is easy to achieve and the blood cells are important in tumor immunity, which changes after RFA. On the one hand, the changes in blood cells identify the immune changes of NSCLC; on the other hand, it provides support and suspicion for the treatment of RFA.
Collapse
Affiliation(s)
- Yunfang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China.,Graduate School of Perking Union Medical College, China Academy of Medical Sciences, Beijing, China
| | - Dongdong Wang
- Minimally Invasive Interventional Therapy Center Department, Qingdao Municipal Hospital, Qingdao, China
| | - Xiaoguang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China.,Graduate School of Perking Union Medical College, China Academy of Medical Sciences, Beijing, China
| |
Collapse
|
185
|
Yvan-Charvet L, Ng LG. Granulopoiesis and Neutrophil Homeostasis: A Metabolic, Daily Balancing Act. Trends Immunol 2020; 40:598-612. [PMID: 31256783 DOI: 10.1016/j.it.2019.05.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023]
Abstract
Granulopoiesis is part of the hematopoietic hierarchic architecture, where hematopoietic stem cells give rise to highly proliferative multipotent and lineage-committed granulocytic progenitor cells that differentiate into unipotent neutrophil progenitors. Given their short lifespan, neutrophils are rapidly cleared from circulation through specialized efferocytic macrophages. Together with an intrinsic clock, these processes contribute to circadian fluctuations, preserving self-tolerance and protection against invading pathogens. However, metabolic perturbation of granulopoiesis and neutrophil homeostasis can result in low-grade chronic inflammation, as observed with aging. During acute pathogenic infections, hematopoiesis can also be switched into emergency mode, which has been recently associated with significant neutrophil functional heterogeneity. This review focuses on a new reassessment of regulatory mechanisms governing neutrophil production, life-cycle, and diversity in health and disease.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France.
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR, Biopolis, Singapore 138648, Singapore; State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, 288 Nanjing Road, Tianjin 300020, China; School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
186
|
Epithelium-derived IL-33 activates mast cells to initiate neutrophil recruitment following corneal injury. Ocul Surf 2020; 18:633-640. [PMID: 32615259 DOI: 10.1016/j.jtos.2020.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE Neutrophils play a critical role in defending against threats such as microbial infection, yet their activation during innate immune response incites collateral damage to healthy tissues. We have previously shown that corneal injury induces mast cells to express the neutrophil chemoattractant CXCL2. Here we delineate the mechanism of injury-induced, non-IgE-mediated mast cell activation at the ocular surface. METHODS Corneal injury was induced by mechanical removal of the epithelium and anterior stroma in mast cell deficient (cKitW-sh) and C57BL/6 mice using Algerbrush II. Corneas were analyzed for frequencies of total CD45+ inflammatory cells, CD11b+Ly6G+ neutrophils, and cKit+FcεR1+ mast cells using flow cytometry. Mast cells were stimulated with different inflammatory factors known to increase during corneal injury (IL-33, IL-1β, IL-36γ, IL-6, SDF1α and Substance P) and assessed for the secretion of β-hexosaminidase, tryptase and CXCL2 using ELISA. IL-33 neutralizing antibody (1 mg/ml) was administered locally for mast cell inhibition in vivo. RESULTS Mast cell deficient mice failed to recruit early neutrophils to the injured corneas. IL-33 stimulation upregulated CXCL2 secretion by mast cells. Corneal injury resulted in amplified expression of IL-33 at the cornea and epithelium was identified as its primary source. Topical neutralization of IL-33 at the ocular surface inhibited mast cell activation, limited neutrophil infiltration, and reduced corneal inflammatory haze, normalizing tissue architecture following ocular injury. CONCLUSIONS These data implicate IL-33 in mast cell activation and early neutrophil recruitment in non-allergic inflammation, suggesting IL-33 as a potential therapeutic target in inflammatory disorders of the ocular surface.
Collapse
|
187
|
Ginefra P, Lorusso G, Vannini N. Innate Immune Cells and Their Contribution to T-Cell-Based Immunotherapy. Int J Mol Sci 2020; 21:ijms21124441. [PMID: 32580431 PMCID: PMC7352556 DOI: 10.3390/ijms21124441] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
In recent years, immunotherapy has become the most promising therapy for a variety of cancer types. The development of immune checkpoint blockade (ICB) therapies, the adoptive transfer of tumor-specific T cells (adoptive cell therapy (ACT)) or the generation of T cells engineered with chimeric antigen receptors (CAR) have been successfully applied to elicit durable immunological responses in cancer patients. However, not all the patients respond to these therapies, leaving a consistent gap of therapeutic improvement that still needs to be filled. The innate immune components of the tumor microenvironment play a pivotal role in the activation and modulation of the adaptive immune response against the tumor. Indeed, several efforts are made to develop strategies aimed to harness innate immune cells in the context of cancer immunotherapy. In this review, we describe the contribution of innate immune cells in T-cell-based cancer immunotherapy and the therapeutic approaches implemented to broaden the efficacy of these therapies in cancer patients.
Collapse
Affiliation(s)
- Pierpaolo Ginefra
- Laboratory of Immunosenescence and Stem Cell Metabolism, Department of Oncology, Ludwig Cancer Institute, University of Lausanne, 1066 Epalinges, Switzerland;
| | - Girieca Lorusso
- Experimental and Translational Oncology, Department of Oncology, Microbiology, Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland;
| | - Nicola Vannini
- Laboratory of Immunosenescence and Stem Cell Metabolism, Department of Oncology, Ludwig Cancer Institute, University of Lausanne, 1066 Epalinges, Switzerland;
- Correspondence:
| |
Collapse
|
188
|
Hao J, Chen J, Wang M, Zhao J, Wang J, Wang X, Li Y, Tang H. Neutrophils, as "Trojan horses", participate in the delivery of therapeutical PLGA nanoparticles into a tumor based on the chemotactic effect. Drug Deliv 2020; 27:1-14. [PMID: 31818156 PMCID: PMC6920705 DOI: 10.1080/10717544.2019.1701141] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Inspired by the fact that leukocytes have innate phagocytic functions and oriented migration capabilities in response to chemoattractants, we have unveiled that endogenous neutrophils as “Trojan horses”, participate in the delivery of nanoparticles in an “in vivo self-armed assembly” manner. Neutrophils were the main population to preferentially sequester the intravenous administrated nanoparticles with an average size of 260 nm. The pre-implantation of CXCL1-laden hydrogels could trigger and induce a targeted signal to attract an influx of neutrophils carrying the therapeutic goods to the desired position. In mouse models of melanoma, the combinatorial regimen of using the PLGA nanoparticles with the CXCL1 hydrogels exhibited superior tumor inhibition capability. This work leveraged the natural phagocytosis of neutrophile and the chemotactic effect of chemokines for targeted delivery. We believe this strategy will improve the therapeutic efficiency of nanoparticle-based delivery systems, especially when the chemokines are implanted at sites of surgical tumor removal, during cancer treatment at the clinic.
Collapse
Affiliation(s)
- Jifu Hao
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Science, Taian, PR China
| | - Junlan Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Meixiang Wang
- Institute of Immunology, Shandong First Medical University & Shandong Academy of Medical Science, Taian, PR China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, PR China
| | - Jing Zhao
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Science, Taian, PR China
| | - Jianze Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Science, Taian, PR China
| | - Xingrong Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Science, Taian, PR China
| | - Yuhong Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School & Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Hua Tang
- Institute of Immunology, Shandong First Medical University & Shandong Academy of Medical Science, Taian, PR China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, PR China
| |
Collapse
|
189
|
Yang L, Liu L, Zhang R, Hong J, Wang Y, Wang J, Zuo J, Zhang J, Chen J, Hao H. IL-8 mediates a positive loop connecting increased neutrophil extracellular traps (NETs) and colorectal cancer liver metastasis. J Cancer 2020; 11:4384-4396. [PMID: 32489457 PMCID: PMC7255375 DOI: 10.7150/jca.44215] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
Host and tumorous inflammation actively affect liver metastasis of colorectal cancer (CRC). Neutrophils have been recognized as one active participant in metastasis procedure, with controversial roles however. Activated neutrophils release extracellular traps (NETs) which are involved in infection and multiple pathological conditions. NETs on cancer metastasis is getting recognized but less elucidated in mechanism. How NETs interact with cancer cells is still largely unknown. In this study, we found that neutrophils from CRC patients, especially those with liver metastatic, underwent remarkably enhanced NETs. Clinically, sera and pathological NETs marker closely correlated with onset of liver metastasis. Through in vivo and in vitro studies, we proved that increased NETs positively contribute to onset of CRC liver metastasis. Digesting NETs with DNase 1 diminished the increased liver metastasis associated with NETs. In detail, NETs trapped CRC cells in liver and exerted no cytotoxicity on tumor cells, but boosted tumorous proliferation and invasion capacity. We further found this enhanced malignancy of trapped CRC cells was due to the elevated tumorous interleukin (IL)-8 expression triggered by NETs. Blocking IL-8 activity effectively abrogated the enhanced proliferation and invasion triggered by NETs. Moreover, overproduced IL-8 in turn activate neutrophils towards NETs formation, thus forming a positive loop optimizing CRC liver metastasis. Collectively, our study propose a novel positive feedback between elevated tumorous IL-8 and NETs to promote CRC liver metastasis, and identify potential strategy against liver metastasis.
Collapse
Affiliation(s)
- Luyu Yang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China.,Caner Metastasis Institute, Fudan University, Shanghai, China
| | - Lu Liu
- Department of Infection Disease, Huashan Hospital, Fudan University, Shanghai, China
| | - Rui Zhang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Hong
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaping Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jieliang Zuo
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jubo Zhang
- Department of Infection Disease, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hankun Hao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
190
|
Abstract
PURPOSE OF REVIEW Although organ transplantation has become the standard life-saving strategy for patients with end-stage organ failure and those with malignancies, effective and safe therapeutic strategies to combat allograft loss remain to be established. With the emerging evidence suggesting the critical role of innate immunity in the mechanism of allograft injury, we summarize the latest understanding of macrophage-neutrophil cross-communication and discuss therapeutic prospects of their targeting in transplant recipients. RECENT FINDINGS Macrophages and neutrophils contribute to the pathogenesis of early peritransplant ischemia-reperfusion injury and subsequent allograft rejection immune cascade, primarily by exacerbating inflammatory response and tissue damage. Noteworthy, recent advances enabled to elucidate multifaceted functions of innate immune cells, which are not only deleterious but may also prove graft-protective. Indeed, the efficacy of macrophage polarizing regimens or macrophage-targeted migration have been recognized to create graft-protective local environment. Moreover, novel molecular mechanisms in the neutrophil function have been identified, such as neutrophil extracellular traps, tissue-repairing capability, crosstalk with macrophages and T cells as well as reverse migration into the circulation. SUMMARY As efficient strategies to manage allograft rejection and improve transplant outcomes are lacking, newly discovered, and therapeutically attractive innate immune cell functions warrant comprehensive preclinical and clinical attention.
Collapse
|
191
|
Zhao Y, Rahmy S, Liu Z, Zhang C, Lu X. Rational targeting of immunosuppressive neutrophils in cancer. Pharmacol Ther 2020; 212:107556. [PMID: 32343986 DOI: 10.1016/j.pharmthera.2020.107556] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
Neutrophils, the most abundant circulating leukocytes in human, play an indispensable role in the innate immune response to microbial infections. However, the contribution of tumor-associated neutrophils (TANs) to cancer progression and tumor immunity has been a matter of debate for decades. A higher neutrophil-to-lymphocyte ratio is associated with adverse overall survival in many solid tumors. Preclinical evidence exists to support both anti-tumor and pro-tumor activities of TANs, and TANs employ diverse mechanisms to influence tumor progression and metastasis. Here, we focus our review on the immunosuppressive mechanism of TANs and highlight how neutrophils can operate to dampen both innate and adaptive immunity to promote tumorigenesis. Here we discuss the intriguing and sometimes controversial connection between TANs and granulocytic/polymorphonuclear myeloid-derived suppressor cells (G/PMN-MDSCs). The molecular mechanisms underlying neutrophils' role in immunosuppression provide potential therapeutic targets for cancer treatment, either as monotherapies or as a part of combinatorial regimens. Therefore, we also highlight a number of neutrophil-targeting approaches that may improve the efficacy of current anticancer therapies, especially cancer immunotherapy. Currently interest is surging in the understanding and targeting of immunosuppressive neutrophils, with the goal of developing novel therapeutic strategies in the battle against cancer.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sharif Rahmy
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Zhongmin Liu
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai 200092, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA; Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
192
|
Bacterial outer membrane vesicles as a platform for biomedical applications: An update. J Control Release 2020; 323:253-268. [PMID: 32333919 DOI: 10.1016/j.jconrel.2020.04.031] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/02/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022]
Abstract
Outer membrane vesicles (OMVs) are produced by Gram-negative bacteria both in vitro and in vivo. OMVs are nano-sized spherical vehicles formed by lipid bilayer membranes and contain multiple parent bacteria-derived components. Based on the presence of bacterial antigens, pathogen-associated molecular patterns (PAMPs), adhesins, various proteins and the vesicle structure, OMVs have been developed for biomedical applications as bacterial vaccines, adjuvants, cancer immunotherapy agents, drug delivery vehicles, and anti-bacteria adhesion agents. In this review, we analyze the contributions of the structure and composition of OMVs to their applications, summarize the methods used to isolate and characterize OMVs, and highlight recent progress and future perspectives of OMVs in biomedical applications.
Collapse
|
193
|
The complexity of neutrophils in health and disease: Focus on cancer. Semin Immunol 2020; 48:101409. [PMID: 32958359 PMCID: PMC7500440 DOI: 10.1016/j.smim.2020.101409] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022]
Abstract
Neutrophils are essential soldiers of the immune response and their role have long been restricted to their activities in defence against microbial infections and during the acute phase of the inflammatory response. However, increasing number of investigations showed that neutrophils are endowed with plasticity and can participate in the orchestration of both innate and adaptive immune responses. Neutrophils have an impact on a broad range of disorders, including infections, chronic inflammations, and cancer. Neutrophils are present in the tumour microenvironment and have been reported to mediate both pro-tumour and anti-tumour responses. Neutrophils can contribute to genetic instability, tumour cell proliferation, angiogenesis and suppression of the anti-tumour immune response. In contrast, neutrophils are reported to mediate anti-tumour resistance by direct killing of tumour cells or by engaging cooperative interactions with other immune cells. Here we discuss the current understandings of neutrophils biology and functions in health and diseases, with a specific focus on their role in cancer biology and their prognostic significance in human cancer.
Collapse
|
194
|
Role of Neutrophils and Myeloid-Derived Suppressor Cells in Glioma Progression and Treatment Resistance. Int J Mol Sci 2020; 21:ijms21061954. [PMID: 32182988 PMCID: PMC7139844 DOI: 10.3390/ijms21061954] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/29/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Recent efforts in brain tumor research have been directed towards the modulation of the immune system for therapeutic interventions. Several human cancers, including gliomas, are infiltrated with immune cell types-including neutrophils and myeloid-derived suppressor cells-that contribute to tumor progression, invasiveness, and treatment resistance. The role of tumor-associated neutrophils and myeloid-derived suppressor cells in cancer biology remains elusive, as these cells can exert a multitude of pro-tumor and antitumor effects. In this review, we provide the current understanding and novel insights on the role of neutrophils and myeloid-derived suppressor cells in glioma progression and treatment resistance, as well as the mechanisms of pleiotropic behaviors in these cells during disease progression, with an emphasis on possible strategies to reprogram these cells towards their antitumor actions.
Collapse
|
195
|
Abstract
HCC (hepatocellular carcinoma) is the second leading cause of cancer deaths worldwide, with several etiologic causes, mostly inflammation-associated. Different inflammatory responses in the liver can be triggered by different etiological agents. The inflammatory process can be resolved or be persistent, depending on the etiology and multiple other factors. Chronic inflammation, tissue remodeling, genetic alterations, and modifications in cellular signaling are considered to be key processes promoting immunosuppression. The progressive immunosuppression leads to the inactivation of anti-tumor immunity involved in HCC carcinogenesis and progression. Tumor cellular processes including DNA damage, necrosis, and ER (endoplasmic reticulum) stress can affect both immune-surveillance and cancer-promoting inflammation, supporting a mutual interdependence. Here, we review the current understanding of how chronic liver injury and inflammation is triggered and sustained, and how inflammation is linked to HCC. The identification of many hepatic microenvironmental inflammatory processes and their effector molecules, has resulted in extensive translational work and promising clinical trials of new immunomodulatory agents.
Collapse
|
196
|
Neubert E, Meyer D, Kruss S, Erpenbeck L. The power from within - understanding the driving forces of neutrophil extracellular trap formation. J Cell Sci 2020; 133:133/5/jcs241075. [PMID: 32156720 DOI: 10.1242/jcs.241075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are one of the most intriguing discoveries in immunological research of the past few years. After their first description in 2004, the number of research articles on how NETs affect immunodefense, and also how they contribute to an ever-growing number of diseases, has skyrocketed. However, tempting as it may seem to plunge into pharmaceutical approaches to tamper with NET formation, our understanding of this complex process is still incomplete. Important concepts such as the context-dependent dual functions of NETs, in that they are both inflammatory and anti-inflammatory, or the major intra- and extracellular forces driving NET formation, are only emerging. In this Review, we summarize key aspects of our current understanding of NET formation (also termed NETosis), emphasize biophysical aspects and focus on three key principles - rearrangement and destabilization of the plasma membrane and the cytoskeleton, alterations and disassembly of the nuclear envelope, and chromatin decondensation as a driving force of intracellular reorganization.
Collapse
Affiliation(s)
- Elsa Neubert
- Department of Dermatology, Venereology and Allergology, University Medical Center, Göttingen University, 37075 Göttingen, Germany.,Institute of Physical Chemistry, Faculty of Chemistry, Göttingen University, 37077 Göttingen, Germany
| | - Daniel Meyer
- Institute of Physical Chemistry, Faculty of Chemistry, Göttingen University, 37077 Göttingen, Germany
| | - Sebastian Kruss
- Institute of Physical Chemistry, Faculty of Chemistry, Göttingen University, 37077 Göttingen, Germany
| | - Luise Erpenbeck
- Department of Dermatology, Venereology and Allergology, University Medical Center, Göttingen University, 37075 Göttingen, Germany
| |
Collapse
|
197
|
Li M, Li S, Zhou H, Tang X, Wu Y, Jiang W, Tian Z, Zhou X, Yang X, Wang Y. Chemotaxis-driven delivery of nano-pathogenoids for complete eradication of tumors post-phototherapy. Nat Commun 2020; 11:1126. [PMID: 32111847 PMCID: PMC7048836 DOI: 10.1038/s41467-020-14963-0] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery. The presence of several biological barriers impede the efficacy of nano-mediated drug delivery for solid cancer therapy. Here, the authors develop a nano-pathogenoid system that targets circulating neutrophils and show that it overcomes these biological barriers and improves tumour targeting and efficacy.
Collapse
Affiliation(s)
- Min Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China.,Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China.,College of Chemistry and Environmental Engineering, Shenzhen University, 518060, Shenzhen, Guangdong, China
| | - Shuya Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Han Zhou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xinfeng Tang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Yi Wu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Wei Jiang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xuechang Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, 518060, Shenzhen, Guangdong, China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, 510006, Guangzhou, China
| | - Yucai Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China. .,Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China.
| |
Collapse
|
198
|
Bonaventura A, Grossi F, Carbone F, Vecchié A, Minetti S, Bardi N, Elia E, Ansaldo AM, Ferrara D, Rijavec E, Dal Bello MG, Rossi G, Biello F, Tagliamento M, Alama A, Coco S, Spallarossa P, Dallegri F, Genova C, Montecucco F. Resistin is associated with overall survival in non-small cell lung cancer patients during nivolumab treatment. Clin Transl Oncol 2020; 22:1603-1610. [PMID: 32048158 DOI: 10.1007/s12094-020-02305-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
|
199
|
Galon J, Bruni D. Tumor Immunology and Tumor Evolution: Intertwined Histories. Immunity 2020; 52:55-81. [PMID: 31940273 DOI: 10.1016/j.immuni.2019.12.018] [Citation(s) in RCA: 343] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/01/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
Cancer is a complex disease whose outcome depends largely on the cross-talk between the tumor and its microenvironment. Here, we review the evolution of the field of tumor immunology and the advances, in lockstep, of our understanding of cancer as a disease. We discuss the involvement of different immune cells at distinct stages of tumor progression and how immune contexture determinants shaping tumor development are being exploited therapeutically. Current clinical stratification schemes focus on the tumor histopathology and the molecular characteristics of the tumor cell. We argue for the importance of revising these stratification systems to include immune parameters so as to address the immediate need for improved prognostic and/or predictive information to guide clinical decisions.
Collapse
Affiliation(s)
- Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Université de Paris; Centre de Recherche des Cordeliers, F-75006 Paris, France.
| | - Daniela Bruni
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Université de Paris; Centre de Recherche des Cordeliers, F-75006 Paris, France
| |
Collapse
|
200
|
Zhang S, Zhou L, Zhang M, Wang Y, Wang M, Du J, Gu W, Kui F, Li J, Geng S, Du G. Berberine Maintains the Neutrophil N1 Phenotype to Reverse Cancer Cell Resistance to Doxorubicin. Front Pharmacol 2020; 10:1658. [PMID: 32063859 PMCID: PMC7000449 DOI: 10.3389/fphar.2019.01658] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023] Open
Abstract
This study explores the contributions of neutrophils to chemotherapeutic resistance and berberine-regulated cancer cell sensitivity to doxorubicin (DOX). In vitro experiments, continuous DOX treatment led to the shift of HL-60 cells to N2 neutrophils and thus induced chemotherapeutic resistance. The combination treatment with DOX and 2 µM berberine resulted in the differentiation of HL-60 cells toward N1 and therefore stimulated HL-60 cell immune clearance. Berberine increased reactive oxygen species (ROS) and decreased autophagy and therefore induced apoptosis in HL-60-N2 cells with morphological changes, but had no effect on cell viability in HL-60-N1 cells. The neutrophil-regulating efficacy of berberine was confirmed in the urethane-induced lung carcinogenic model and H22 liver cancer allograft model. Furthermore, we found that DOX-derived neutrophils had high levels of CD133 and CD309 surface expression, which prevented both chemotherapeutic sensitivity and immune rejection by self-expression of PD-L1 and surface expression of PD-1 receptor on T cells, whereas berberine could downregulate CD133 and CD309 surface expression. Finally, berberine-relevant targets and pathways were evaluated. This study first suggests an important role of berberine in regulating neutrophil phenotypes to maintain cancer cell sensitivity to DOX.
Collapse
Affiliation(s)
- Shuhui Zhang
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Lin Zhou
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Mengdi Zhang
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Yuehua Wang
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Mengqi Wang
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Jincheng Du
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
- Chinese Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Wenwen Gu
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Fuguang Kui
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Jiahuan Li
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
| | - Shengnan Geng
- School of Pharmacy and Chemical Engineering, Zhengzhou University of Industry Technology, Xinzheng, China
| | - Gangjun Du
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, China
- School of Pharmacy and Chemical Engineering, Zhengzhou University of Industry Technology, Xinzheng, China
| |
Collapse
|