1
|
Zhao M, Chen S, Xu J, Zhou F, Zhou M, Tian S, Ding Z, Chen Y. Alleviation of sepsis-induced lung and liver injury by polysaccharides from Tetrastigma hemsleyanum Diels et Gilg via suppression of TLR4/NF-κB/COX-2 pathway and modulation of immune checkpoint molecules. Mol Immunol 2025; 179:52-64. [PMID: 39919350 DOI: 10.1016/j.molimm.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/06/2024] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
Sepsis, a common clinical complication, leads to multi-organ damage due to systemic infection and currently lacks effective therapeutic drugs. Polysaccharide derived from Tetrastigma hemsleyanum Diels et Gilg (TH), abbreviated as THP, is a water-soluble component extracted from TH, exhibiting anti-inflammatory and immunomodulatory properties. This study aims to investigate the effects and mechanisms of THP in sepsis. Results demonstrated that THP reduced neutrophils in the peripheral blood of mice established by cecal ligation and puncture (CLP), and inhibited IL-6 and MCP-1 in plasma, thereby improving systemic inflammation. THP ameliorated pulmonary edema, mitigated lung and liver histopathological injuries, reduced infiltration of neutrophils and macrophages in the lung and liver, and inhibited the TNF-α, IL-6, IL-1β, and MCP-1 transcription in both organs. Additionally, THP decreased myeloid cells, neutrophils, monocytes, and Tregs in the spleens of septic mice, while increasing T cells, CD4+ T cells, and CD8+ T cells, thereby restoring immune imbalance. Mechanistically, THP attenuated sepsis by inhibiting the overactivation of the TLR4/NF-κB/COX-2 pathway, and reducing PD-1, PD-L1, IDO1 in the lung, and PD-1, PD-L1 in the liver of septic mice. In conclusion, this study provides theoretical support for the potential application of THP in sepsis treatment.
Collapse
Affiliation(s)
- Mengjia Zhao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310053, China; School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Senmiao Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jingwen Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310053, China; School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fangmei Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310053, China; School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mingyuan Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310053, China; School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shasha Tian
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhishan Ding
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310053, China; School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yuchi Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310053, China; School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
2
|
Kumari S, Biswas A, Maiti TK, Bandyopadhyay B, Banerjee A. Induction of PD-1 and CD44 in CD4 + T cells by circulatory extracellular vesicles from severe dengue patients drives endothelial damage via the NF-kB signaling pathway. J Virol 2025; 99:e0186124. [PMID: 39745465 PMCID: PMC11852895 DOI: 10.1128/jvi.01861-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/13/2024] [Indexed: 02/26/2025] Open
Abstract
Extracellular vesicles (EVs) emerged as critical contributors to the pathogenesis of vascular endothelial barrier dysfunction during the inflammatory response to infection. However, the contribution of circulating EVs to modifying endothelial function during dengue virus infection remains unclear. In this study, we showed that severe dengue patients' plasma-derived EV (SD-EV) were found to carry elevated levels of different protein cargos, e.g., immunoregulatory proteins (PD-L1, CD44). Further, we demonstrated that SD-EV induces PD-1 and CD44 expression on CD4+ T cells. SD-EV-modulated CD4+ T (SD-EV-CD4) cells released secretome delayed endothelial cell (EC) migration, arrested them in the G1 phase, and augmented the expression of PD-L1 and ICAM-1 expression on EC through the Notch signaling pathway. Blocking SD-EV and CD4+ T-cell interaction through the PD-1/PD-L1 pathway partially rescued the CD4+ T cell's effect on EC but did not alter ICAM-1 expression on EC. We observed that the ICAM-1 expression on EC and hyaluronic acid (HA) release from EC was mediated by CD44, which was elevated on SD-EV-modulated CD4+ T cells (SD-EV-CD4), indicating a permeability defect. Blocking of CD44 on SD-EV-CD4 significantly reduced ICAM-1 expression on EC. Further, depletion of specific cytokines, e.g., TNF-α and not IFN-γ from the SD-EV-CD4 secretome, reduced ICAM-1 expression, decreased transendothelial electrical resistance, and induced apoptosis on EC significantly. Treatment with NF-kB inhibitor before secretome addition to EC reduced ICAM-1 expression on EC. In conclusion, we provided evidence that SD-EV-CD4 carrying PD-1 and CD44, when interacting with EC, significantly affected endothelial cell properties and may be significant in dengue-mediated endothelial dysfunction.IMPORTANCEExtracellular vesicles (EVs) are small membrane vesicles secreted into biological fluids, including plasma from living cells, holding insights into pathological processes. Studying EVs under pathological conditions is extremely important as they play a selective role in intercellular communication and modulation of immune response under diverse pathological conditions. However, there is less clarity on how circulatory extracellular vesicles influence immune cells during dengue virus (DV) infection and impact pathogenesis. Our present study highlights the impact of severe dengue patients' plasma-derived EV (SD-EV) on CD4+ T cells and together induce endothelial barrier dysfunction. We provided evidence that SD-EV induces PD-1 and CD44 on CD4+ T cells and, when interacting with endothelial cells (EC), drives endothelial damage through direct interaction or secretome and may be significant in dengue-mediated endothelial dysfunction.
Collapse
Affiliation(s)
- Sharda Kumari
- Laboratory of Virology, Regional Centre for Biotechnology, National Capital Region Biotechnology Science Cluster, Faridabad, Haryana, India
| | - Ankit Biswas
- Functional Proteomics Laboratory, Regional Centre for Biotechnology, National Capital Region Biotechnology Science Cluster, Faridabad, Haryana, India
| | - Tushar Kanti Maiti
- Functional Proteomics Laboratory, Regional Centre for Biotechnology, National Capital Region Biotechnology Science Cluster, Faridabad, Haryana, India
| | | | - Arup Banerjee
- Laboratory of Virology, Regional Centre for Biotechnology, National Capital Region Biotechnology Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
3
|
An L, Shao R, Hang C, Wang X, Zhang L, Cui H, Yu J, Shan Z, Tang Z. Monocyte programmed death-ligand 1 upregulation in early post-out-of-hospital cardiac arrest is associated with increased risk of acute respiratory distress syndrome. Resusc Plus 2024; 20:100822. [PMID: 39559732 PMCID: PMC11570934 DOI: 10.1016/j.resplu.2024.100822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Background Out-of-hospital cardiac arrest (OHCA) is a major public health problem. Acute respiratory distress syndrome (ARDS) is a common condition in OHCA patients. We investigated the relationship between the expression of programmed death-1 (PD-1) related molecules and the development and prognosis of ARDS. Methods Between January 2021 and December 2023, post-resuscitated patients were screened for eligibility in the study. PD-1 related molecules expression was measured by flow cytometry at 48 h of admission in patients with OHCA. The prognostic variables were the development of ARDS during hospitalization and the 28-day patient mortality rate. We analyzed the relationship between the expression of PD-1-related molecules and the development of secondary ARDS in OHCA patients, and assessed the correlation of this expression with the prognosis of ARDS patients. Results In total, 107 consecutive OHCA patients were enrolled in this study. The median age of the enrolled patients was 60 years, with an age range of 53 to 67 years, and 71 % were male. Among the cardiac arrest patients, 44.8 % had a cardiac etiology, 30.8 % were witnessed, 17.8 % received bystander CPR, and 66.4 % had an initial rhythm of asystole. Our results showed that only monocyte ligand programmed death ligand-1 (PD-L1) expression was significantly elevated in the ARDS group of OHCA patients (P < 0.001). Among patients with ARDS, the expression of PD-L1 on monocytes in non-survivors was significantly higher than in survivors (P < 0.05). The Receiver operating characteristic curves analysis demonstrates that monocyte PD-L1 expression has predictive potential for the development and prognosis of ARDS. Multivariate logistic regression analysis showed that monocyte PD-L1 expression was an independent predictor of mortality in OHCA patients with ARDS. Conclusions This study indicates that patients with increased PD-L1 on monocytes after OHCA may be more likely to develop ARDS. The expression of PD-L1 on monocytes was an independent predictive factor for the incidence of ARDS and mortality rate in OHCA patients.
Collapse
Affiliation(s)
- Le An
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Rui Shao
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Chenchen Hang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xingsheng Wang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Luying Zhang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hao Cui
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jingfei Yu
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhenyu Shan
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Ziren Tang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing 100020, China
| |
Collapse
|
4
|
Tao T, Zhu Y, Shi Y, Sun B, Gu Y, Xu S. Unveiling the role of PD-L1 in vascular endothelial dysfunction: Insights into the mtros/NLRP3/caspase-1 mediated pyroptotic pathway. Exp Cell Res 2024; 438:114047. [PMID: 38631546 DOI: 10.1016/j.yexcr.2024.114047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Programmed death ligand-1(PD-L1) has been postulated to play a crucial role in the regulation of barrier functions of the vascular endothelium, yet how this novel molecule mediates dysfunction in endothelial cells (ECs) during acute lung injury (ALI) remains largely unknown. METHODS PD-L1 siRNA and plasmids were synthesized and applied respectively to down- or up-regulate PD-L1 expression in human lung microvascular endothelial cells (HMVECs). RNA sequencing was used to explore the differentially expressed genes following PD-L1 overexpression. The expression levels of tight junction proteins (ZO-1 and occludin) and the signaling pathways of NLRP-3/caspase-1/pyroptosis were analyzed. A mouse model of indirect ALI was established through hemorrhagic shock (HEM) followed by cecal ligation and puncture (CLP), enabling further investigation into the effects of intravenous delivery of PD-L1 siRNA. RESULTS A total of 1502 differentially expressed genes were identified, comprising 532 down-regulated and 970 up-regulated genes in ECs exhibiting PD-L1overexpression. Enrichment of PD-L1-correlated genes were observed in the NOD-like receptor signaling pathway and the TNF signaling pathway. Western blot assays confirmed that PD-L1 overexpression elevated the expression of NLRP3, cleaved-caspase-1, ASC and GSDMD, and concurrently diminished the expression of ZO-1 and occludin. This overexpression also enhanced mitochondrial oxidative phosphorylation and mitochondrial reactive oxygen species (mtROS) production. Interestingly, mitigating mitochondrial dysfunction with mitoQ partially countered the adverse effects of PD-L1 on the functionality of ECs. Furthermore, intravenous administration of PD-L1 siRNA effectively inhibited the activation of the NLRP3 inflammasome and pyroptosis in pulmonary ECs, subsequently ameliorating lung injury in HEM/CLP mice. CONCLUSION PD-L1-mediated activation of the inflammasome contributes significantly to the disruption of tight junction and induction of pyroptosis in ECs, where oxidative stress associated with mitochondrial dysfunction serves as a pivotal mechanism underpinning these effects.
Collapse
Affiliation(s)
- Tianzhu Tao
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
| | - Ying Zhu
- Department of Pulmonary and Critical Care Medicine, 7th Medical Center of Chinese PLA General Hospital, Beijing, 100700, China; College of Pulmonary and Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Yue Shi
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
| | - Bingke Sun
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Gu
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumin Xu
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Tang N, Yang Y, Xie Y, Yang G, Wang Q, Li C, Liu Z, Huang JA. CD274 (PD-L1) negatively regulates M1 macrophage polarization in ALI/ARDS. Front Immunol 2024; 15:1344805. [PMID: 38440722 PMCID: PMC10909908 DOI: 10.3389/fimmu.2024.1344805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Background Acute lung injury (ALI)/severe acute respiratory distress syndrome (ARDS) is a serious clinical syndrome characterized by a high mortality rate. The pathophysiological mechanisms underlying ALI/ARDS remain incompletely understood. Considering the crucial role of immune infiltration and macrophage polarization in the pathogenesis of ALI/ARDS, this study aims to identify key genes associated with both ALI/ARDS and M1 macrophage polarization, employing a combination of bioinformatics and experimental approaches. The findings could potentially reveal novel biomarkers for the diagnosis and management of ALI/ARDS. Methods Gene expression profiles relevant to ALI were retrieved from the GEO database to identify co-upregulated differentially expressed genes (DEGs). GO and KEGG analyses facilitated functional annotation and pathway elucidation. PPI networks were constructed to identify hub genes, and differences in immune cell infiltration were subsequently examined. The expression of hub genes in M1 versus M2 macrophages was evaluated using macrophage polarization datasets. The diagnostic utility of CD274 (PD-L1) for ARDS was assessed by receiver operating characteristic (ROC) analysis in a validation dataset. Experimental confirmation was conducted using two LPS-induced M1 macrophage models and an ALI mouse model. The role of CD274 (PD-L1) in M1 macrophage polarization and associated proinflammatory cytokine production was further investigated by siRNA-mediated silencing. Results A total of 99 co-upregulated DEGs were identified in two ALI-linked datasets. Enrichment analysis revealed that these DEGs were mainly involved in immune-inflammatory pathways. The following top 10 hub genes were identified from the PPI network: IL-6, IL-1β, CXCL10, CD274, CCL2, TLR2, CXCL1, CCL3, IFIT1, and IFIT3. Immune infiltration analysis revealed a significantly increased abundance of M1 and M2 macrophages in lung tissue from the ALI group compared to the control group. Subsequent analysis confirmed that CD274 (PD-L1), a key immunological checkpoint molecule, was highly expressed within M1 macrophages. ROC analysis validated CD274 (PD-L1) as a promising biomarker for the diagnosis of ARDS. Both in vitro and in vivo experiments supported the bioinformatics analysis and confirmed that the JAK-STAT3 pathway promotes CD274 (PD-L1) expression on M1 macrophages. Importantly, knockdown of CD274 (PD-L1) expression potentiated M1 macrophage polarization and enhanced proinflammatory cytokines production. Conclusion This study demonstrates a significant correlation between CD274 (PD-L1) and M1 macrophages in ALI/ARDS. CD274 (PD-L1) functions as a negative regulator of M1 polarization and the secretion of proinflammatory cytokines in macrophages. These findings suggest potential new targets for the diagnosis and treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Nana Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yang Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Yifei Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Guohui Yang
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qin Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Chang Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Jian-an Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, China
| |
Collapse
|
6
|
Lacinski RA, Dziadowicz SA, Stewart A, Chaharbakhshi E, Akhter H, Pisquiy JJ, Victory JH, Hardham JB, Chew C, Prorock A, Bao Y, Sol-Church K, Hobbs GR, Klein E, Nalesnik MA, Hu G, de Oliveira A, Santiago SP, Lindsey BA. Nanosphere pharmacodynamics improves safety of immunostimulatory cytokine therapy. iScience 2024; 27:108836. [PMID: 38303687 PMCID: PMC10831265 DOI: 10.1016/j.isci.2024.108836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/04/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Systemic administration of interleukin (IL)-12 induces potent anti-tumor immune responses in preclinical cancer models through the systemic activation of effector immune cells and release of proinflammatory cytokines. IL-12-loaded PLGA nanospheres (IL12ns) are hypothesized to improve therapeutic efficacy and thwart unwanted side effects observed in previous human clinical trials. Through the investigation of peripheral blood and local tissue immune responses in healthy BALB/c mice, the immune-protective pharmacodynamics of IL12ns were suggested. Nanospheres increased pro-inflammatory plasma cytokines/chemokines (IFN-γ, IL-6, TNF-α, and CXCL10) without inducing maladaptive transcriptomic signatures in circulating peripheral immune cells. Gene expression profiling revealed activation of pro-inflammatory signaling pathways in systemic tissues, the likely source of these effector cytokines. These data support that nanosphere pharmacodynamics, including shielding IL-12 from circulating immune cells, depositing peripherally in systemic immune tissues, and then slowly eluting bioactive cytokine, thereafter, are essential to safe immunostimulatory therapy.
Collapse
Affiliation(s)
- Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Amanda Stewart
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Chaharbakhshi
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - John J. Pisquiy
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Jack H. Victory
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Joshua B. Hardham
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Claude Chew
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alyson Prorock
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Yongde Bao
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Katia Sol-Church
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Gerald R. Hobbs
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Michael A. Nalesnik
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Ana de Oliveira
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Stell P. Santiago
- Department of Pathology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Brock A. Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
7
|
Chen Y, Guo DZ, Zhu CL, Ren SC, Sun CY, Wang Y, Wang JF. The implication of targeting PD-1:PD-L1 pathway in treating sepsis through immunostimulatory and anti-inflammatory pathways. Front Immunol 2023; 14:1323797. [PMID: 38193090 PMCID: PMC10773890 DOI: 10.3389/fimmu.2023.1323797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Sepsis currently remains a major contributor to mortality in the intensive care unit (ICU), with 48.9 million cases reported globally and a mortality rate of 22.5% in 2017, accounting for almost 20% of all-cause mortality worldwide. This highlights the urgent need to improve the understanding and treatment of this condition. Sepsis is now recognized as a dysregulation of the host immune response to infection, characterized by an excessive inflammatory response and immune paralysis. This dysregulation leads to secondary infections, multiple organ dysfunction syndrome (MODS), and ultimately death. PD-L1, a co-inhibitory molecule expressed in immune cells, has emerged as a critical factor in sepsis. Numerous studies have found a significant association between the expression of PD-1/PD-L1 and sepsis, with a particular focus on PD-L1 expressed on neutrophils recently. This review explores the role of PD-1/PD-L1 in immunostimulatory and anti-inflammatory pathways, illustrates the intricate link between PD-1/PD-L1 and sepsis, and summarizes current therapeutic approaches against PD-1/PD-L1 in the treatment and prognosis of sepsis in preclinical and clinical studies.
Collapse
Affiliation(s)
- Yu Chen
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - De-zhi Guo
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - Cheng-long Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shi-chun Ren
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chen-yan Sun
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jia-feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
8
|
Battaglini D, Iavarone IG, Al-Husinat L, Ball L, Robba C, Silva PL, Cruz FF, Rocco PR. Anti-inflammatory therapies for acute respiratory distress syndrome. Expert Opin Investig Drugs 2023; 32:1143-1155. [PMID: 37996088 DOI: 10.1080/13543784.2023.2288080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/22/2023] [Indexed: 11/25/2023]
Abstract
INTRODUCTION Treatments for the acute respiratory distress syndrome (ARDS) are mainly supportive, and ventilatory management represents a key approach in these patients. Despite progress in pharmacotherapy, anti-inflammatory strategies for the treatment of ARDS have shown controversial results. Positive outcomes with pharmacologic and nonpharmacologic treatments have been found in two different biological subphenotypes of ARDS, suggesting that, with a personalized medicine approach, pharmacotherapy for ARDS can be effective. AREAS COVERED This article reviews the literature concerning anti-inflammatory therapies for ARDS, focusing on pharmacological and stem-cell therapies, including extracellular vesicles. EXPERT OPINION Despite advances, ARDS treatments remain primarily supportive. Ventilatory and fluid management are important strategies in these patients that have demonstrated significant impacts on outcome. Anti-inflammatory drugs have shown some benefits, primarily in preclinical research and in specific clinical scenarios, but no recommendations are available from guidelines to support their use in patients with ARDS, except in particular settings such as different subphenotypes, specific etiologies, or clinical trials. Personalized medicine seems promising insofar as it may identify specific subgroups of patients with ARDS who may benefit from anti-inflammatory treatment. However, additional efforts are needed to move subphenotype characterization from bench to bedside.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Ida Giorgia Iavarone
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Lou'i Al-Husinat
- Department of Clinical Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Lorenzo Ball
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Chiara Robba
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rm Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Chong L, Ahmadvand N, Noori A, Lv Y, Chen C, Bellusci S, Zhang JS. Injury activated alveolar progenitors (IAAPs): the underdog of lung repair. Cell Mol Life Sci 2023; 80:145. [PMID: 37166489 PMCID: PMC10173924 DOI: 10.1007/s00018-023-04789-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023]
Abstract
Alveolar epithelial type II cells (AT2s) together with AT1s constitute the epithelial lining of lung alveoli. In contrast to the large flat AT1s, AT2s are cuboidal and smaller. In addition to surfactant production, AT2s also serve as prime alveolar progenitors in homeostasis and play an important role during regeneration/repair. Based on different lineage tracing strategies in mice and single-cell transcriptomic analysis, recent reports highlight the heterogeneous nature of AT2s. These studies present compelling evidence for the presence of stable or transitory AT2 subpopulations with distinct marker expression, signaling pathway activation and functional properties. Despite demonstrated progenitor potentials of AT2s in maintaining homeostasis, through self-renewal and differentiation to AT1s, the exact identity, full progenitor potential and regulation of these progenitor cells, especially in the context of human diseases remain unclear. We recently identified a novel subset of AT2 progenitors named "Injury-Activated Alveolar Progenitors" (IAAPs), which express low levels of Sftpc, Sftpb, Sftpa1, Fgfr2b and Etv5, but are highly enriched for the expression of the surface receptor programmed cell death-ligand 1 (Pd-l1). IAAPs are quiescent during lung homeostasis but activated upon injury with the potential to proliferate and differentiate into AT2s. Significantly, a similar population of PD-L1 positive cells expressing intermediate levels of SFTPC are found to be expanded in human IPF lungs. We summarize here the current understanding of this newly discovered AT2 progenitor subpopulation and also try to reconcile the relationship between different AT2 stem cell subpopulations regarding their progenitor potential, regulation, and relevance to disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Lei Chong
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Negah Ahmadvand
- Department of Cell Biology, Duke University School of Medicine, Durham, NC27710, USA
| | - Afshin Noori
- Cardio Pulmonary Institute, Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center, Justus-Liebig University Giessen, 35392, Giessen, Germany
| | - Yuqing Lv
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China
| | - Chengshui Chen
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology and Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Saverio Bellusci
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China.
- Laboratory of Extracellular Matrix Remodelling, Cardio Pulmonary Institute, Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center, Member of the German Lung Center, Justus-Liebig University Giessen, 35392, Giessen, Germany.
| | - Jin-San Zhang
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology and Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
10
|
Li Y, Tan R, Li R, Tian R, Liu Z, Wang X, Chen E, Pan T, Qu H. PKM2/STAT1-mediated PD-L1 upregulation on neutrophils during sepsis promotes neutrophil organ accumulation by serving an anti-apoptotic role. J Inflamm (Lond) 2023; 20:16. [PMID: 37131151 PMCID: PMC10155438 DOI: 10.1186/s12950-023-00341-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/25/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Delayed neutrophil apoptosis during sepsis may impact neutrophil organ accumulation and tissue immune homeostasis. Elucidating the mechanisms underlying neutrophil apoptosis may help identify potential therapeutic targets. Glycolysis is critical to neutrophil activities during sepsis. However, the precise mechanisms through which glycolysis regulates neutrophil physiology remain under-explored, especially those involving the non-metabolic functions of glycolytic enzymes. In the present study, the impact of programmed death ligand-1 (PD-L1) on neutrophil apoptosis was explored. The regulatory effect of the glycolytic enzyme, pyruvate kinase M2 (PKM2), whose role in septic neutrophils remains unaddressed, on neutrophil PD-L1 expression was also explored. METHODS Peripheral blood neutrophils were isolated from patients with sepsis and healthy controls. PD-L1 and PKM2 levels were determined by flow cytometry and Western blotting, respectively. Dimethyl sulfoxide (DMSO)-differentiated HL-60 cells were stimulated with lipopolysaccharide (LPS) as an in vitro simulation of septic neutrophils. Cell apoptosis was assessed by annexin V/propidium iodide (annexin V/PI) staining, as well as determination of protein levels of cleaved caspase-3 and myeloid cell leukemia-1 (Mcl-1) by Western blotting. An in vivo model of sepsis was constructed by intraperitoneal injection of LPS (5 mg/kg) for 16 h. Pulmonary and hepatic neutrophil infiltration was assessed by flow cytometry or immunohistochemistry. RESULTS PD-L1 level was elevated on neutrophils under septic conditions. Administration of neutralizing antibodies against PD-L1 partially reversed the inhibitory effect of LPS on neutrophil apoptosis. Neutrophil infiltration into the lung and liver was also reduced in PD-L1-/- mice 16 h after sepsis induction. PKM2 was upregulated in septic neutrophils and promoted neutrophil PD-L1 expression both in vitro and in vivo. In addition, PKM2 nuclear translocation was increased after LPS stimulation, which promoted PD-L1 expression by directly interacting with and activating signal transducer and activator of transcription 1 (STAT1). Inhibition of PKM2 activity or STAT1 activation also led to increased neutrophil apoptosis. CONCLUSION In this study, a PKM2/STAT1-mediated upregulation of PD-L1 on neutrophils and the anti-apoptotic effect of upregulated PD-L1 on neutrophils during sepsis were identified, which may result in increased pulmonary and hepatic neutrophil accumulation. These findings suggest that PKM2 and PD-L1 could serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Yinjiaozhi Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Erzhen Chen
- Department of Emergency Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
11
|
Armstead BE, Lee CS, Chen Y, Zhao R, Chung CS, Fredericks AM, Monaghan SF, Ayala A. Application of single cell multiomics points to changes in chromatin accessibility near calcitonin receptor like receptor and a possible role for adrenomedullin in the post-shock lung. Front Med (Lausanne) 2023; 10:1003121. [PMID: 37113606 PMCID: PMC10126233 DOI: 10.3389/fmed.2023.1003121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a commonly occurring sequelae of traumatic injury resulting from indirect insults like hypovolemic shock and/or extrapulmonary sepsis. The high lethality rate associated with these pathologies outlines the importance of clarifying the "priming" effects seen in the post-shock lung microenvironment, which are understood to bring about a dysregulated or overt immune response when triggered by a secondary systemic infectious/septic challenge culminating in ALI. In this pilot project, we test the hypothesis that application of a single cell multiomics approach can elucidate novel phenotype specific pathways potentially contributing to shock-induced ALI/ARDS. Methods Hypovolemic shock was induced in C57BL/6 (wild-type), PD-1, PD-L1, or VISTA gene deficient male mice, 8-12 weeks old. Wild-type sham surgeries function as negative controls. A total of 24-h post-shock rodents were sacrificed, their lungs harvested and sectioned, with pools prepared from 2 mice per background, and flash frozen on liquid nitrogen. N = 2 biological replicates (representing 4 mice total) were achieved for all treatment groups across genetic backgrounds. Samples were received by the Boas Center for Genomics and Human Genetics, where single cell multiomics libraries were prepared for RNA/ATAC sequencing. The analysis pipeline Cell Ranger ARC was implemented to attain feature linkage assessments across genes of interest. Results Sham (pre-shock) results suggest high chromatin accessibility around calcitonin receptor like receptor (CALCRL) across cellular phenotypes with 17 and 18 feature links, exhibiting positive correlation with gene expression between biological replicates. Similarity between both sample chromatin profiles/linkage arcs is evident. Post-shock wild-type accessibility is starkly reduced across replicates where the number of feature links drops to 1 and 3, again presenting similar replicate profiles. Samples from shocked gene deficient backgrounds displayed high accessibility and similar profiles to the pre-shock lung microenvironment. Conclusion High pre-shock availability of DNA segments and their positive correlation with CALCRL gene expression suggests an apparent regulatory capacity on transcription. Post-shock gene deficient chromatin profiles presented similar results to that of pre-shock wild-type samples, suggesting an influence on CALCRL accessibility. Key changes illustrated in the pre-ALI context of shock may allow for additional resolution of "priming" and "cellular pre-activation/pre-disposition" processes within the lung microenvironment.
Collapse
Affiliation(s)
- Brandon E. Armstead
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
| | - Chung Sunny Lee
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Yaping Chen
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Runping Zhao
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Chun-Shiang Chung
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Alger M. Fredericks
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- The Miriam Hospital, Providence, RI, United States
- The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Sean F. Monaghan
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
- The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Alfred Ayala
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
- The Warren Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
12
|
Zhang T, Yu-Jing L, Ma T. Role of regulation of PD-1 and PD-L1 expression in sepsis. Front Immunol 2023; 14:1029438. [PMID: 36969168 PMCID: PMC10035551 DOI: 10.3389/fimmu.2023.1029438] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Long term immunosuppression is problematic during sepsis. The PD-1 and PD-L1 immune checkpoint proteins have potent immunosuppressive functions. Recent studies have revealed several features of PD-1 and PD-L1 and their roles in sepsis. Here, we summarize the overall findings of PD-1 and PD-L1 by first reviewing the biological features of PD-1 and PD-L1 and then discussing the mechanisms that control the expression of PD-1 and PD-L1. We then review the functions of PD-1 and PD-L1 in physiological settings and further discuss PD-1 and PD-L1 in sepsis, including their involvement in several sepsis-related processes and their potential therapeutic relevance in sepsis. In general, PD-1 and PD-L1 have critical roles in sepsis, indicating that their regulation may be a potential therapeutic target for sepsis.
Collapse
Affiliation(s)
- Teng Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Yu-Jing
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Tao Ma,
| |
Collapse
|
13
|
Ravaglia C, Doglioni C, Chilosi M, Piciucchi S, Dubini A, Rossi G, Pedica F, Puglisi S, Donati L, Tomassetti S, Poletti V. Clinical, radiological and pathological findings in patients with persistent lung disease following SARS-CoV-2 infection. Eur Respir J 2022; 60:2102411. [PMID: 35301248 PMCID: PMC8932282 DOI: 10.1183/13993003.02411-2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 02/13/2022] [Indexed: 12/18/2022]
Abstract
Some patients experience pulmonary sequelae after SARS-CoV-2 infection, ranging from self-limited abnormalities to major lung diseases. Morphological analysis of lung tissue may help our understanding of pathogenic mechanisms and help to provide consistent personalised management. The aim of this study was to ascertain morphological and immunomolecular features of lung tissue. Transbronchial lung cryobiopsy was carried out in patients with persistent symptoms and computed tomography suggestive of residual lung disease after recovery from SARS-CoV-2 infection. 164 patients were referred for suspected pulmonary sequelae after COVID-19; 10 patients with >5% parenchymal lung disease underwent lung biopsy. The histological pattern of lung disease was not homogeneous and three different case clusters could be identified, which was mirrored by their clinical and radiological features. Cluster 1 ("chronic fibrosing") was characterised by post-infection progression of pre-existing interstitial pneumonias. Cluster 2 ("acute/subacute injury") was characterised by different types and grades of lung injury, ranging from organising pneumonia and fibrosing nonspecific interstitial pneumonia to diffuse alveolar damage. Cluster 3 ("vascular changes") was characterised by diffuse vascular increase, dilatation and distortion (capillaries and venules) within otherwise normal parenchyma. Clusters 2 and 3 had immunophenotypical changes similar to those observed in early/mild COVID-19 pneumonias (abnormal expression of STAT3 in hyperplastic pneumocytes and PD-L1, IDO and STAT3 in endothelial cells). This is the first study correlating histological/immunohistochemical patterns with clinical and radiological pictures of patients with post-COVID lung disease. Different phenotypes with potentially different underlying pathogenic mechanisms have been identified.
Collapse
Affiliation(s)
- Claudia Ravaglia
- Dept of Thoracic Diseases, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Claudio Doglioni
- Dept of Pathology, University Vita-Salute, Milan and San Raffaele Scientific Institute, Milan, Italy
| | - Marco Chilosi
- Dept of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Sara Piciucchi
- Dept of Radiology, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Alessandra Dubini
- Dept of Pathology, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Giulio Rossi
- Dept of Pathology, Fondazione Poliambulanza Istituto Ospedaliero Multispecialistico, Brescia, Italy
| | - Federica Pedica
- Dept of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Puglisi
- Dept of Thoracic Diseases, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Luca Donati
- Biostatistics and Clinical Trial Unit, Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori"-IRST S.r.l., IRCCS, Meldola, Italy
| | - Sara Tomassetti
- Dept of Experimental and Clinical Medicine, Careggi University Hospital, Firenze, Italy
| | - Venerino Poletti
- Dept of Thoracic Diseases, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
- DIMES, University of Bologna, Bologna, Italy
- Dept of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
14
|
Huang Q, Gao S, Yao Y, Wang Y, Li J, Chen J, guo C, Zhao D, Li X. Innate immunity and immunotherapy for hemorrhagic shock. Front Immunol 2022; 13:918380. [PMID: 36091025 PMCID: PMC9453212 DOI: 10.3389/fimmu.2022.918380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022] Open
Abstract
Hemorrhagic shock (HS) is a shock result of hypovolemic injury, in which the innate immune response plays a central role in the pathophysiology ofthe severe complications and organ injury in surviving patients. During the development of HS, innate immunity acts as the first line of defense, mediating a rapid response to pathogens or danger signals through pattern recognition receptors. The early and exaggerated activation of innate immunity, which is widespread in patients with HS, results in systemic inflammation, cytokine storm, and excessive activation of complement factors and innate immune cells, comprised of type II innate lymphoid cells, CD4+ T cells, natural killer cells, eosinophils, basophils, macrophages, neutrophils, and dendritic cells. Recently, compelling evidence focusing on the innate immune regulation in preclinical and clinical studies promises new treatment avenues to reverse or minimize HS-induced tissue injury, organ dysfunction, and ultimately mortality. In this review, we first discuss the innate immune response involved in HS injury, and then systematically detail the cutting-edge therapeutic strategies in the past decade regarding the innate immune regulation in this field; these strategies include the use of mesenchymal stem cells, exosomes, genetic approaches, antibody therapy, small molecule inhibitors, natural medicine, mesenteric lymph drainage, vagus nerve stimulation, hormones, glycoproteins, and others. We also reviewed the available clinical studies on immune regulation for treating HS and assessed the potential of immune regulation concerning a translation from basic research to clinical practice. Combining therapeutic strategies with an improved understanding of how the innate immune system responds to HS could help to identify and develop targeted therapeutic modalities that mitigate severe organ dysfunction, improve patient outcomes, and reduce mortality due to HS injury.
Collapse
Affiliation(s)
- Qingxia Huang
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Song Gao
- Jilin Xiuzheng Pharmaceutical New Drug Development Co., Ltd., Changchun, China
| | - Yao Yao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yisa Wang
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jinjin Chen
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chen guo
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Daqing Zhao, ; Xiangyan Li,
| | - Xiangyan Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Daqing Zhao, ; Xiangyan Li,
| |
Collapse
|
15
|
Dexmedetomidine Attenuates LPS-Stimulated Alveolar Type II Cells’ Injury through Upregulation of miR-140-3p and Partial Suppression of PD-L1 Involving Inactivating JNK-Bnip3 Pathway. Can Respir J 2022; 2022:8433960. [PMID: 35958433 PMCID: PMC9357803 DOI: 10.1155/2022/8433960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/10/2022] [Accepted: 07/14/2022] [Indexed: 11/24/2022] Open
Abstract
Dexmedetomidine (DEX), which is reported to be a newly discovered, novel α-2 adrenoceptor agonist, is known to exhibit anti-inflammatory properties in several diseases. DEX regulates inflammation-related signaling pathways and genes through interactions with several miRNAs. This study verified that expression levels of miR-140-3p were diminished when alveolar type II cells were exposed to LPS. However, the levels of miR-140-3p were confirmed as showing an increase with DEX treatment. These observations revealed that the expression of miR-140-3p was related to the beneficial effects that accompanied the DEX treatment of LPS-induced ALI. In addition, PD-1/PD-L1 expression increased extensively when RLE-6TN cells were induced by LPS. The increased expression was reduced after treatment with DEX. Thus, it appears that the PD-L1 expression was targeted directly by miR-140-3p, resulting in the partial repression of PD-L1 levels, which involved the inhibition of p-JNK and Bnip3 expression. Therefore, DEX was shown to inhibit the PD-L1 expression by promoting partially increased miR-140-3p levels in RLE-6TN cells. DEX also inactivated the JNK-Bnip3 pathway, resulting in the inhibition of inflammation and alleviating alveolar type II cell injury.
Collapse
|
16
|
Piao W, Li L, Saxena V, Iyyathurai J, Lakhan R, Zhang Y, Lape IT, Paluskievicz C, Hippen KL, Lee Y, Silverman E, Shirkey MW, Riella LV, Blazar BR, Bromberg JS. PD-L1 signaling selectively regulates T cell lymphatic transendothelial migration. Nat Commun 2022; 13:2176. [PMID: 35449134 PMCID: PMC9023578 DOI: 10.1038/s41467-022-29930-0] [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: 07/27/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
Programmed death-1 (PD-1) and its ligand PD-L1 are checkpoint molecules which regulate immune responses. Little is known about their functions in T cell migration and there are contradictory data about their roles in regulatory T cell (Treg) function. Here we show activated Tregs and CD4 effector T cells (Teffs) use PD-1/PD-L1 and CD80/PD-L1, respectively, to regulate transendothelial migration across lymphatic endothelial cells (LECs). Antibody blockade of Treg PD-1, Teff CD80 (the alternative ligand for PD-L1), or LEC PD-L1 impairs Treg or Teff migration in vitro and in vivo. PD-1/PD-L1 signals through PI3K/Akt and ERK to regulate zipper junctional VE-cadherin, and through NFκB-p65 to up-regulate VCAM-1 expression on LECs. CD80/PD-L1 signaling up-regulates VCAM-1 through ERK and NFκB-p65. PD-1 and CD80 blockade reduces tumor egress of PD-1high fragile Tregs and Teffs into draining lymph nodes, respectively, and promotes tumor regression. These data provide roles for PD-L1 in cell migration and immune regulation.
Collapse
Affiliation(s)
- Wenji Piao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jegan Iyyathurai
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ram Lakhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yigang Zhang
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN, 55455, USA
| | - Isadora Tadeval Lape
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, Boston, MA, 02114, USA
| | - Christina Paluskievicz
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Keli L Hippen
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN, 55455, USA
| | - Young Lee
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Emma Silverman
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Marina W Shirkey
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Leonardo V Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, Boston, MA, 02114, USA
| | - Bruce R Blazar
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN, 55455, USA
| | - Jonathan S Bromberg
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
17
|
Zhu J, Li J, Chung CS, Lomas-Neira JL, Ayala A. Patho-Mechanisms for Hemorrhage/Sepsis-Induced Indirect Acute Respiratory Distress Syndrome: A Role for Lung TIE1 and Its Regulation by Neutrophils. Shock 2022; 57:608-615. [PMID: 34907117 PMCID: PMC8916968 DOI: 10.1097/shk.0000000000001902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Severe hemorrhage (Hem) has been shown to be causal for the development of extra-pulmonary/indirect acute respiratory distress syndrome (iARDS) and is associated with severe endothelial cell (EC) injury. EC growth factors, Angiopoietin (Ang)-1 and -2, maintain vascular homeostasis via tightly regulated competitive interaction with the tyrosine kinase receptor, Tie2, expressed on ECs. OBJECTIVE Since it has been reported that the orphan receptor, Tie1, may be able to play a role in Ang:Tie2 signaling; we chose to examine Tie1's capacity to alter the lung Ang:Tie2 interaction in response to the sequential insults of shock/sepsis (cecal ligation and puncture [CLP]), culminating in iARDS. METHODS Male mice were subjected to Hem alone or sequential Hem followed 24 hours later by CLP that induces iARDS. Changes in lung and/or plasma levels of Tie1, Tie2, Ang-1, Ang-2, various systemic cytokine/chemokines and indices of lung injury/inflammation were then determined. The role of Tie1 was established by intravenous administration of Tie1 specific or control siRNA at 1 h post-Hem. Alternatively, the contribution of neutrophils was assessed by pre-treating mice with anti-neutrophil antibody depletion 48 h prior to Hem. RESULTS Lung tissue levels of Tie1 expression elevated over the first 6 to 24 h post-Hem alone. Subsequently, we found that treatment of Hem/CLP mice with Tie1-specific siRNA not only decreased Tie1 expression in lung tissue compared to control siRNA, but, suppressed the rise in lung inflammatory cytokines, lung MPO and the rise in lung protein leak. Finally, much as we have previously shown that neutrophil interaction with resident pulmonary vascular ECs contribute significantly to Ang-2 release and EC dysfunction, central to the development of iARDS. Here, we report that depletion of neutrophils also decreased lung tissue Tie1 expression and increased Tie2 activation in Hem/CLP mice. CONCLUSION Together, these data imply that shock-induced increased expression of Tie1 can contribute to EC activation by inhibiting Ang:Tie2 interaction, culminating in EC dysfunction and the development of iARDS.
Collapse
Affiliation(s)
- Jiali Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital & the Alpert School of Medicine at Brown University, Providence, RI 02830, USA
| | - Jinbao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital & the Alpert School of Medicine at Brown University, Providence, RI 02830, USA
| | - Joanne L. Lomas-Neira
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital & the Alpert School of Medicine at Brown University, Providence, RI 02830, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital & the Alpert School of Medicine at Brown University, Providence, RI 02830, USA
| |
Collapse
|
18
|
Gao X, Li W, Syed F, Yuan F, Li P, Yu Q. PD-L1 signaling in reactive astrocytes counteracts neuroinflammation and ameliorates neuronal damage after traumatic brain injury. J Neuroinflammation 2022; 19:43. [PMID: 35135580 PMCID: PMC8822654 DOI: 10.1186/s12974-022-02398-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tissue damage and cellular destruction are the major events in traumatic brain injury (TBI), which trigger sterile neuroimmune and neuroinflammatory responses in the brain. While appropriate acute and transient neuroimmune and neuroinflammatory responses facilitate the repair and adaptation of injured brain tissues, prolonged and excessive neuroimmune and neuroinflammatory responses exacerbate brain damage. The mechanisms that control the intensity and duration of neuroimmune and neuroinflammatory responses in TBI largely remain elusive. METHODS We used the controlled cortical impact (CCI) model of TBI to study the role of immune checkpoints (ICPs), key regulators of immune homeostasis, in the regulation of neuroimmune and neuroinflammatory responses in the brain in vivo. RESULTS We found that de novo expression of PD-L1, a potent inhibitory ICP, was robustly and transiently induced in reactive astrocytes, but not in microglia, neurons, or oligodendrocyte progenitor cells (OPCs). These PD-L1+ reactive astrocytes were highly enriched to form a dense zone around the TBI lesion. Blockade of PD-L1 signaling enlarged brain tissue cavity size, increased infiltration of inflammatory Ly-6CHigh monocytes/macrophages (M/Mɸ) but not tissue-repairing Ly-6CLowF4/80+ M/Mɸ, and worsened TBI outcomes in mice. PD-L1 gene knockout enhanced production of CCL2 that is best known for its ability to interact with its cognate receptor CCR2 on Ly-6CHigh M/Mϕ to chemotactically recruit these cells into inflammatory sites. Mechanically, PD-L1 signaling in astrocytes likely exhibits dual inhibitory activities for the prevention of excessive neuroimmune and neuroinflammatory responses to TBI through (1) the PD-1/PD-L1 axis to suppress the activity of brain-infiltrating PD-1+ immune cells, such as PD-1+ T cells, and (2) PD-L1 intrinsic signaling to regulate the timing and intensity of astrocyte reactions to TBI. CONCLUSIONS PD-L1+ astrocytes act as a gatekeeper to the brain to control TBI-related neuroimmune and neuroinflammatory responses, thereby opening a novel avenue to study the role of ICP-neuroimmune axes in the pathophysiology of TBI and other neurological disorders.
Collapse
Affiliation(s)
- Xiang Gao
- Spinal Cord and Brain Injury Research Group, Department of Neurological Surgery, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN, 46202, USA.
| | - Wei Li
- Department of Microbiology and Immunology, Medical Science Building, MS267, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Fahim Syed
- Department of Microbiology and Immunology, Medical Science Building, MS267, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Fang Yuan
- Spinal Cord and Brain Injury Research Group, Department of Neurological Surgery, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN, 46202, USA
| | - Ping Li
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Qigui Yu
- Department of Microbiology and Immunology, Medical Science Building, MS267, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA.
| |
Collapse
|
19
|
Silva RCMC, Panis C, Pires BRB. Lessons from transmissible cancers for immunotherapy and transplant. Immunol Med 2021; 45:146-161. [PMID: 34962854 DOI: 10.1080/25785826.2021.2018783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The emergence of horizontal transmission of cancer between vertebrates is an issue that interests scientists and medical society. Transmission requires: (i) a mechanism by which cancer cells can transfer to another organism and (ii) a repressed immune response on the part of the recipient. Transmissible tumors are unique models to comprehend the responses and mechanisms mediated by the major histocompatibility complex (MHC), which can be transposed for transplant biology. Here, we discuss the mechanisms involved in immune-mediated tissue rejection, making a parallel with transmissible cancers. We also discuss cellular and molecular mechanisms involved in cancer immunotherapy and anti-rejection therapies.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio De Janeiro, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná, UNIOESTE, Francisco Beltrão, Brazil
| | | |
Collapse
|
20
|
Kong L, Deng J, Zhou X, Cai B, Zhang B, Chen X, Chen Z, Wang W. Sitagliptin activates the p62-Keap1-Nrf2 signalling pathway to alleviate oxidative stress and excessive autophagy in severe acute pancreatitis-related acute lung injury. Cell Death Dis 2021; 12:928. [PMID: 34635643 PMCID: PMC8505515 DOI: 10.1038/s41419-021-04227-0] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022]
Abstract
Acute lung injury (ALI) is a complication of severe acute pancreatitis (SAP). Sitagliptin (SIT) is a DPP4 inhibitor that exerts anti-inflammatory and antioxidant effects; however, its mechanism of action in SAP-ALI remains unclear. In this study, we investigated the effects of SIT on SAP-ALI and the specific pathways involved in SAP-induced lung inflammation, including oxidative stress, autophagy, and p62-Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) signalling pathways. Nrf2 knockout (Nrf2-/-) and wild-type (WT) mice were pre-treated with SIT (100 mg/kg), followed by caerulein and lipopolysaccharide (LPS) administration to induce pancreatic and lung injury. BEAS-2B cells were transfected with siRNA-Nrf2 and treated with LPS, and the changes in inflammation, reactive oxygen species (ROS) levels, and autophagy were measured. SIT reduced histological damage, oedema, and myeloperoxidase activity in the lung, decreased the expression of pro-inflammatory cytokines, and inhibited excessive autophagy and ROS production via the activation of the p62-Keap1-Nrf2 signalling pathway and promotion of the nuclear translocation of Nrf2. In Nrf2-knockout mice, the anti-inflammatory effect of SIT was reduced, resulting in ROS accumulation and excessive autophagy. In BEAS-2B cells, LPS induced ROS production and activated autophagy, further enhanced by Nrf2 knockdown. This study demonstrates that SIT reduces SAP-ALI-associated oxidative stress and excessive autophagy through the p62-Keap1-Nrf2 signalling pathway and nuclear translocation of Nrf2, suggesting its therapeutic potential in SAP-ALI.
Collapse
Affiliation(s)
- Lingming Kong
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Deng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiang Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Binbin Cai
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Baofu Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaohu Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zongjing Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Weiming Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
21
|
Chilosi M, Poletti V, Ravaglia C, Rossi G, Dubini A, Piciucchi S, Pedica F, Bronte V, Pizzolo G, Martignoni G, Doglioni C. The pathogenic role of epithelial and endothelial cells in early-phase COVID-19 pneumonia: victims and partners in crime. Mod Pathol 2021; 34:1444-1455. [PMID: 33883694 PMCID: PMC8058579 DOI: 10.1038/s41379-021-00808-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 02/04/2023]
Abstract
Current understanding of the complex pathogenesis of COVID-19 interstitial pneumonia pathogenesis in the light of biopsies carried out in early/moderate phase and histology data obtained at postmortem analysis is discussed. In autopsies the most observed pattern is diffuse alveolar damage with alveolar-epithelial type-II cell hyperplasia, hyaline membranes, and frequent thromboembolic disease. However, these observations cannot explain some clinical, radiological and physiopathological features observed in SARS-CoV-2 interstitial pneumonia, including the occurrence of vascular enlargement on CT and preserved lung compliance in subjects even presenting with or developing respiratory failure. Histological investigation on early-phase pneumonia on perioperative samples and lung biopsies revealed peculiar morphological and morpho-phenotypical changes including hyper-expression of phosphorylated STAT3 and immune checkpoint molecules (PD-L1 and IDO) in alveolar-epithelial and endothelial cells. These features might explain in part these discrepancies.
Collapse
Affiliation(s)
- Marco Chilosi
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy.
| | - Venerino Poletti
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
- Department of Diseases of the Thorax, G.B. Morgagni Hospital, Forlì, Italy
| | - Claudia Ravaglia
- Department of Diseases of the Thorax, G.B. Morgagni Hospital, Forlì, Italy
| | - Giulio Rossi
- Department of Pathology, Ravenna Hospital, Ravenna, Italy
| | | | - Sara Piciucchi
- Department of Radiology, G.B. Morgagni Hospital, Forlì, Italy
| | - Federica Pedica
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Vincenzo Bronte
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Giovanni Pizzolo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Claudio Doglioni
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Cross AR, Lion J, Poussin K, Glotz D, Mooney N. Inflammation Determines the Capacity of Allogenic Endothelial Cells to Regulate Human Treg Expansion. Front Immunol 2021; 12:666531. [PMID: 34305898 PMCID: PMC8299527 DOI: 10.3389/fimmu.2021.666531] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/23/2021] [Indexed: 11/25/2022] Open
Abstract
During allotransplantation, the endothelium acts as semi-professional antigen-presenting cells with the ability to activate proliferation and to promote differentiation of CD4+-T subsets. These abilities are dependent on the luminal expression of HLA class II antigens by microvascular endothelial cells, which is regulated by inflammatory cytokines. The upregulation of HLA-DR and HLA-DQ during rejection implies significant intragraft inflammation. Furthermore, the microvascular inflammation is an independent determinant for renal allograft failure. In this study, the potential of inflammation to modify endothelial regulation of peripheral CD4+ Treg cells was examined. Microvascular endothelial cells were exposed to pro-inflammatory cytokines for varying durations before co-culture with PBMC from non-HLA matched donors. Proliferation and expansion of CD4+Treg and soluble factor secretion was determined. Early interactions were detected by phosphorylation of Akt. Video microscopy was used to examine spatial and temporal endothelial-CD4+T interactions. Highly inflammatory conditions led to increased endothelial expression of HLA-DR, the adhesion molecule ICAM-1, the costimulatory molecule PD-L1 and de novo expression of HLA-DQ. Treg differentiation was impaired by exposure of endothelial cells to a high level of inflammation. Neither IL-6, IL-2 nor TGFβ were implicated in reducing Treg numbers. High PD-L1 expression interfered with early endothelial cell interactions with CD4+T lymphocytes and led to modified TCR signaling. Blocking endothelial PD-L1 resulted in a partial restoration of Treg. The allogenic endothelial cell-mediated expansion of Treg depends on a critical threshold of inflammation. Manipulation of the PD-L1/PD-1 pathway or endothelial activation post-transplantation may promote or interfere with this intrinsic mechanism of allospecific Treg expansion.
Collapse
Affiliation(s)
- Amy Rachael Cross
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U976, Paris, France.,Université de Paris, INSERM U976, Paris, France
| | - Julien Lion
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U976, Paris, France
| | - Karine Poussin
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U976, Paris, France
| | - Denis Glotz
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U976, Paris, France.,Université de Paris, INSERM U976, Paris, France.,Service de Néphrologie et Transplantation, Hôpital Saint Louis, Paris, France
| | - Nuala Mooney
- Human Immunology, Pathophysiology and Immunotherapy, INSERM U976, Paris, France.,Université de Paris, INSERM U976, Paris, France
| |
Collapse
|
23
|
Zhao S, Gao J, Li J, Wang S, Yuan C, Liu Q. PD-L1 Regulates Inflammation in LPS-Induced Lung Epithelial Cells and Vascular Endothelial Cells by Interacting with the HIF-1α Signaling Pathway. Inflammation 2021; 44:1969-1981. [PMID: 34014492 DOI: 10.1007/s10753-021-01474-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/31/2022]
Abstract
Sepsis-induced lung injury was the most common cause of death in patients. This study aimed to investigate whether PD-L1 regulates the inflammation in LPS-induced lung epithelial cells and vascular endothelial cells by interacting with the HIF-1α signaling pathway. Sepsis-induced lung injury mice were constructed by cecal ligation and puncture (CLP) procedure, and lipopolysaccharide (LPS)-induced lung epithelial cells and vascular endothelial cells simulate the sepsis-induced lung injury model in vitro. Hematoxylin-eosin (HE) staining detected the morphological changes of the lung tissues, and immunohistochemistry (IHC) detected the PD-L1 expression in lung tissues. Bicinchoninic acid (BCA) assay determined the protein concentration in bronchial alveolar lavage fluid (BALF). The number of PD-1 (+) cells in blood was detected by flow cytometry. The apoptosis in lung tissues and LPS-induced cells was analyzed by TUNEL assay. The inflammatory factor levels and HIF-1α in lung tissues and LPS-induced cells were analyzed by ELISA. The transfection effects of KD-PDL1 or KD-HIF1A in lung epithelial cells and vascular endothelial cells were confirmed by qRT-PCR analysis. The protein expression related to the PD-L1- and HIF-1α-related pathway was determined by Western blot analysis. As a result, LMT-28, as an IL-6 inhibitor, alleviated lung injury and suppressed the apoptosis and inflammation in lung tissues in BALF and the number of PD-1 (+) cells in blood. Sepsis-induced lung injury activated the PD-L1- and HIF-1α-related pathway, while LMT-28 could not completely inhibit the pathway. In addition, downregulation of PD-L1 or downregulation of HIF-1α suppressed the apoptosis and alleviated the inflammation in LPS-induced lung epithelial cells and vascular endothelial cells. Downregulation of PD-L1 had significant effects on lung epithelial cells but had greater effects on vascular endothelial cells. Downregulation of HIF-1α could decrease PD-L1 expression, and downregulation of PD-L1 could also suppress the protein expression of HIF-1α and related pathways. In conclusion, downregulation of PD-L1 alleviated the inflammation in LPS-induced lung epithelial cells and vascular endothelial cells by suppressing the HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Shilong Zhao
- Department of Respiratory Intensive Care Unit (ICU), The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe East Road, Zhengzhou, 450000, Henan Province, China
| | - Jing Gao
- Department of Respiratory Intensive Care Unit (ICU), The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe East Road, Zhengzhou, 450000, Henan Province, China
| | - Jing Li
- Department of Respiratory Intensive Care Unit (ICU), The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe East Road, Zhengzhou, 450000, Henan Province, China
| | - Shilei Wang
- Department of Respiratory Intensive Care Unit (ICU), The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe East Road, Zhengzhou, 450000, Henan Province, China
| | - Congcong Yuan
- Department of Respiratory Intensive Care Unit (ICU), The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe East Road, Zhengzhou, 450000, Henan Province, China
| | - Qiuhong Liu
- Department of Respiratory Intensive Care Unit (ICU), The First Affiliated Hospital of Zhengzhou University, No. 1 JianShe East Road, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
24
|
Degauque N, Haziot A, Brouard S, Mooney N. Endothelial cell, myeloid, and adaptive immune responses in SARS-CoV-2 infection. FASEB J 2021; 35:e21577. [PMID: 33831263 PMCID: PMC8250117 DOI: 10.1096/fj.202100024r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is an emerging respiratory pathogen that has rapidly spread in human populations. Severe forms of infection associate cytokine release syndrome and acute lung injury due to hyperinflammatory responses even though virus clearance is achieved. Key components of inflammation include immune cell recruitment in infected tissues, a step which is under the control of endothelial cells. Here, we review endothelial cell responses in inflammation and infection due to SARS-CoV-2 together with phenotypic and functional alterations of monocytes, T and B lymphocytes with which they interact. We surmise that endothelial cells function as an integrative and active platform for the various cells recruited, where fine tuning of immune responses takes place and which provides opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Nicolas Degauque
- Centre De Recherche En Transplantation Et ImmunologieUMR1064, INSERM, Université De NantesNantesFrance
- Institut De Transplantation Urologie Néphrologie (ITUN)CHU NantesNantesFrance
- Laboratoire d’ImmunologieCHU NantesNantesFrance
| | - Alain Haziot
- INSERM U976Institut de Recherche Saint LouisParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| | - Sophie Brouard
- Centre De Recherche En Transplantation Et ImmunologieUMR1064, INSERM, Université De NantesNantesFrance
- Institut De Transplantation Urologie Néphrologie (ITUN)CHU NantesNantesFrance
- Laboratoire d’ImmunologieCHU NantesNantesFrance
| | - Nuala Mooney
- INSERM U976Institut de Recherche Saint LouisParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| |
Collapse
|
25
|
Concentrated Secretome of Adipose Stromal Cells Limits Influenza A Virus-Induced Lung Injury in Mice. Cells 2021; 10:cells10040720. [PMID: 33804896 PMCID: PMC8063825 DOI: 10.3390/cells10040720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Despite vaccination and antivirals, influenza remains a communicable disease of high burden, with limited therapeutic options available to patients that develop complications. Here, we report the development and preclinical characterization of Adipose Stromal Cell (ASC) concentrated secretome (CS), generated by process adaptable to current Good Manufacturing Practices (cGMP) standards. We demonstrate that ASC-CS limits pulmonary histopathological changes, infiltration of inflammatory cells, protein leak, water accumulation, and arterial oxygen saturation (spO2) reduction in murine model of lung infection with influenza A virus (IAV) when first administered six days post-infection. The ability to limit lung injury is sustained in ASC-CS preparations stored at −80 °C for three years. Priming of the ASC with inflammatory factors TNFα and IFNγ enhances ASC-CS ability to suppress lung injury. IAV infection is associated with dramatic increases in programmed cell death ligand (PDL1) and angiopoietin 2 (Angpt2) levels. ASC-CS application significantly reduces both PDL1 and Angpt2 levels. Neutralization of PDL1 with anti-mouse PDL1 antibody starting Day6 onward effectively ablates lung PDL1, but only non-significantly reduces Angpt2 release. Most importantly, late-phase PDL1 neutralization results in negligible suppression of protein leakage and inflammatory cell infiltration, suggesting that suppression of PDL1 does not play a critical role in ASC-CS therapeutic effects.
Collapse
|
26
|
Fallon EA, Chung CS, Heffernan DS, Chen Y, De Paepe ME, Ayala A. Survival and Pulmonary Injury After Neonatal Sepsis: PD1/PDL1's Contributions to Mouse and Human Immunopathology. Front Immunol 2021; 12:634529. [PMID: 33746973 PMCID: PMC7965961 DOI: 10.3389/fimmu.2021.634529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Morbidity and mortality associated with neonatal sepsis remains a healthcare crisis. PD1-/- neonatal mice endured experimental sepsis, in the form of cecal slurry (CS), and showed improved rates of survival compared to wildtype (WT) counterparts. End-organ injury, particularly of the lung, contributes to the devastation set forth by neonatal sepsis. PDL1-/- neonatal mice, in contrast to PD1-/- neonatal mice did not have a significant improvement in survival after CS. Because of this, we focused subsequent studies on the impact of PD1 gene deficiency on lung injury. Here, we observed that at 24 h post-CS (but not at 4 or 12 h) there was a marked increase in pulmonary edema (PE), neutrophil influx, myeloperoxidase (MPO) levels, and cytokine expression sham (Sh) WT mice. Regarding pulmonary endothelial cell (EC) adhesion molecule expression, we observed that Zona occludens-1 (ZO-1) within the cell shifted from a membranous location to a peri-nuclear location after CS in WT murine cultured ECs at 24hrs, but remained membranous among PD1-/- lungs. To expand the scope of this inquiry, we investigated human neonatal lung tissue. We observed that the lungs of human newborns exposed to intrauterine infection had significantly higher numbers of PD1+ cells compared to specimens who died from non-infectious causes. Together, these data suggest that PD1/PDL1, a pathway typically thought to govern adaptive immune processes in adult animals, can modulate the largely innate neonatal pulmonary immune response to experimental septic insult. The potential future significance of this area of study includes that PD1/PDL1 checkpoint proteins may be viable therapeutic targets in the septic neonate.
Collapse
Affiliation(s)
- Eleanor A. Fallon
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Daithi S. Heffernan
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
- Department of Surgery, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Monique E. De Paepe
- Department of Pathology, Women & Infants Hospital and Alpert Medical School of Brown University, Providence, RI, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
27
|
McBride MA, Patil TK, Bohannon JK, Hernandez A, Sherwood ER, Patil NK. Immune Checkpoints: Novel Therapeutic Targets to Attenuate Sepsis-Induced Immunosuppression. Front Immunol 2021; 11:624272. [PMID: 33613563 PMCID: PMC7886986 DOI: 10.3389/fimmu.2020.624272] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a leading cause of death in intensive care units and survivors develop prolonged immunosuppression and a high incidence of recurrent infections. No definitive therapy exists to treat sepsis and physicians rely on supportive care including antibiotics, intravenous fluids, and vasopressors. With the rising incidence of antibiotic resistant microbes, it is becoming increasingly critical to discover novel therapeutics. Sepsis-induced leukocyte dysfunction and immunosuppression is recognized as an important contributor towards increased morbidity and mortality. Pre-clinical and clinical studies show that specific cell surface inhibitory immune checkpoint receptors and ligands including PD-1, PD-L1, CTLA4, BTLA, TIM3, OX40, and 2B4 play important roles in the pathophysiology of sepsis by mediating a fine balance between host immune competency and immunosuppression. Pre-clinical studies targeting the inhibitory effects of these immune checkpoints have demonstrated reversal of leukocyte dysfunction and improved host resistance of infection. Measurement of immune checkpoint expression on peripheral blood leukocytes may serve as a means of stratifying patients to direct individualized therapy. This review focuses on advances in our understanding of the role of immune checkpoints in the host response to infections, and the potential clinical application of therapeutics targeting the inhibitory immune checkpoint pathways for the management of septic patients.
Collapse
Affiliation(s)
- Margaret A. McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tazeen K. Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Julia K. Bohannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Edward R. Sherwood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Naeem K. Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
28
|
Delicate Role of PD-L1/PD-1 Axis in Blood Vessel Inflammatory Diseases: Current Insight and Future Significance. Int J Mol Sci 2020; 21:ijms21218159. [PMID: 33142805 PMCID: PMC7663405 DOI: 10.3390/ijms21218159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint molecules are the antigen-independent generator of secondary signals that aid in maintaining the homeostasis of the immune system. The programmed death ligand-1 (PD-L1)/PD-1 axis is one among the most extensively studied immune-inhibitory checkpoint molecules, which delivers a negative signal for T cell activation by binding to the PD-1 receptor. The general attributes of PD-L1's immune-suppressive qualities and novel mechanisms on the barrier functions of vascular endothelium to regulate blood vessel-related inflammatory diseases are concisely reviewed. Though targeting the PD-1/PD-L1 axis has received immense recognition-the Nobel Prize in clinical oncology was awarded in the year 2018 for this discovery-the use of therapeutic modulating strategies for the PD-L1/PD-1 pathway in chronic inflammatory blood vessel diseases is still limited to experimental models. However, studies using clinical specimens that support the role of PD-1 and PD-L1 in patients with underlying atherosclerosis are also detailed. Of note, delicate balances in the expression levels of PD-L1 that are needed to preserve T cell immunity and to curtail acute as well as chronic infections in underlying blood vessel diseases are discussed. A significant link exists between altered lipid and glucose metabolism in different cells and the expression of PD-1/PD-L1 molecules, and its possible implications on vascular inflammation are justified. This review summarizes the most recent insights concerning the role of the PD-L1/PD-1 axis in vascular inflammation and, in addition, provides an overview exploring the novel therapeutic approaches and challenges of manipulating these immune checkpoint proteins, PD-1 and PD-L1, for suppressing blood vessel inflammation.
Collapse
|
29
|
Yu Y, Sun H, Zhu L, Ji L, Liu H. Downregulating lncRNA PRNCR1 ameliorates LPS-induced pulmonary vascular endothelial cell injury by modulating miR-330-5p/TLR4 axis. J Biochem Mol Toxicol 2020; 35:e22644. [PMID: 33049095 DOI: 10.1002/jbt.22644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/08/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary vascular endothelial cell (PVEC) injury following acute lung injury or acute respiratory distress syndrome seriously affects disease development. Recently, accumulating evidence has suggested that long noncoding RNA (lncRNA) exerts significant effects in vascular endothelial cell injury. However, PRNCR1, a novel lncRNA, remains scarcely understood in terms of its functions in PVEC injury. Both in vivo and in vitro models of PVEC injury were constructed by lipopolysaccharide (LPS) administration. The relative expressions of PRNCR1, miR-330-5p, and TLR4 were detected by quantitative reverse transcription-polymerase chain reaction, Western blot, and immunohistochemistry. Besides, gain and loss assays of PRNCR1/miR-330-5p were conducted to verify their effects on LPS-induced PVEC injury. Cell Counting Kit-8 assay used to measure cell viability and flow cytometry was used to detect apoptosis. Besides, the protein levels of caspase 3, nuclear factor-κB (NF-κB), and inflammatory cytokines (including tumor necrosis factor-α, interleukin-1β [IL-1β], and IL-6) were evaluated via Western blot and enzyme-linked immunosorbent assay. Moreover, a dual-luciferase activity experiment and RNA immunoprecipitation were applied to confirm the targeting relationship between PRNCR1 and miR-330-5p, miR-330-5p, and TLR4. PRNCR1 and TLR4 levels were significantly upregulated in LPS-treated PVEC, both in vivo and in vitro, while miR-330-5p were downregulated. Inhibiting PRNCR1 or overexpressing miR-330-5p markedly attenuated LPS-induced PVEC injury, expressions of TLR4, NF-κB, and inflammatory cytokines. Mechanistically, PRNCR1 functioned as a competitive endogenous RNA by sponging miR-330-5p and then promoting TLR4 expression. PRNCR1 was upregulated in LPS-induced PVEC and aggravated its injury via modulating the miR-330-5p/TLR4 axis.
Collapse
Affiliation(s)
- Yingqing Yu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongzhi Sun
- Department of Intensive Medicine, Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Zhu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Lianfeng Ji
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Haibo Liu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
30
|
Chen J, Zhang X, Xie J, Xue M, Liu L, Yang Y, Qiu H. Overexpression of TGFβ1 in murine mesenchymal stem cells improves lung inflammation by impacting the Th17/Treg balance in LPS-induced ARDS mice. Stem Cell Res Ther 2020; 11:311. [PMID: 32698911 PMCID: PMC7374869 DOI: 10.1186/s13287-020-01826-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 12/26/2022] Open
Abstract
Background T helper 17 cells (Th17)/regulatory T cells (Treg), as subtypes of CD4+ T cells, play an important role in the inflammatory response of acute respiratory distress syndrome (ARDS). However, there is still a lack of effective methods to regulate the differentiation balance of Th17/Treg. It was proven that mesenchymal stem cells (MSCs) could regulate the differentiation of CD4+ T cells, but the mechanism is still unclear. TGFβ1, a paracrine cytokine of MSCs, could also regulate the differentiation of Th17/Treg but is lowly expressed in MSCs. Therefore, mouse MSCs (mMSCs) overexpressing TGFβ1 were constructed by lentivirus transduction and intratracheally transplanted into LPS-induced ARDS mice in our study. The aim of this study was to evaluate the therapeutic effects of mMSCs overexpressing TGFβ1 on inflammation and immunoregulation by impacting the Th17/Treg balance in LPS-induced ARDS mice. Methods mMSCs overexpressing TGFβ1 were constructed using lentiviral vectors. Then, mouse bone-marrow-derived MSCs (mBM-MSC) and mBM-MSC-TGFβ1 (mBM-MSC overexpressing TGFβ1) were transplanted intratracheally into ARDS mice induced by lipopolysaccharide. At 3 and 7 days after transplantation, the mice were sacrificed, and the homing of the mMSCs was assayed by ex vivo optical imaging. The relative numbers of Th17 and Treg in the lungs and spleens of mice were detected by FCM. IL-17A and IL-10 levels in the lungs of mice were analysed by western blot. Permeability and inflammatory cytokines were evaluated by analysing the protein concentration of BALF using ELISA. Histopathology of the lungs was assessed by haematoxylin and eosin staining and lung injury scoring. Alveolar lung fibrosis was assessed by Masson’s trichrome staining and Ashcroft scoring. The mortality of ARDS mice was followed until 7 days after transplantation. Results The transduction efficiencies mediated by the lentiviral vectors ranged from 82.3 to 88.6%. Overexpressing TGFβ1 inhibited the proliferation of mMSCs during days 5–7 (p < 0.05) but had no effect on mMSC differentiation or migration (p > 0.05). Compared to that in the LPS + mBM-MSC-NC group mice, engraftment of mMSCs overexpressing TGFβ1 led to much more differentiation of T cells into Th17 or Treg (p < 0.05), improved permeability of injured lungs (p < 0.05) and ameliorative histopathology of lung tissue in ARDS mice (p < 0.05). Moreover, IL-17A content was also decreased while IL-10 content was increased in the LPS + mBM-MSC-TGFβ1 group compared with those in the LPS + mBM-MSC-NC group (p < 0.05). Finally, mMSCs overexpressing TGFβ1 did not aggravate lung fibrosis in ARDS mice (p > 0.05). Conclusion MSCs overexpressing TGFβ1 could regulate lung inflammation and attenuate lung injuries by modulating the imbalance of Th17/Treg in the lungs of ARDS mice.
Collapse
Affiliation(s)
- Jianxiao Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China
| | - Xiwen Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China
| | - Jianfeng Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China
| | - Ming Xue
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China
| | - Ling Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
31
|
anlotinib alters tumor immune microenvironment by downregulating PD-L1 expression on vascular endothelial cells. Cell Death Dis 2020; 11:309. [PMID: 32366856 PMCID: PMC7198575 DOI: 10.1038/s41419-020-2511-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022]
Abstract
Aberrant vascular network is a hallmark of cancer. However, the role of vascular endothelial cells (VECs)-expressing PD-L1 in tumor immune microenvironment and antiangiogenic therapy remains unclear. In this study, we used the specimens of cancer patients for immunohistochemical staining to observe the number of PD-L1+ CD34+ VECs and infiltrated immune cells inside tumor specimens. Immunofluorescence staining and flow cytometry were performed to observe the infiltration of CD8+ T cells and FoxP3+ T cells in tumor tissues. Here, we found that PD-L1 expression on VECs determined CD8+ T cells’, FoxP3+ T cells’ infiltration, and the prognosis of patients with lung adenocarcinoma. Anlotinib downregulated PD-L1 expression on VECs through the inactivation of AKT pathway, thereby improving the ratio of CD8/FoxP3 inside tumor and remolding the immune microenvironment. In conclusion, our results demonstrate that PD-L1 high expression on VECs inhibits the infiltration of CD8+ T cells, whereas promotes the aggregation of FoxP3+ T cells into tumor tissues, thus becoming an “immunosuppressive barrier”. Anlotinib can ameliorate the immuno-microenvironment by downregulating PD-L1 expression on VECs to inhibit tumor growth.
Collapse
|
32
|
Xu S, Yang Q, Bai J, Tao T, Tang L, Chen Y, Chung CS, Fallon EA, Ayala A. Blockade of endothelial, but not epithelial, cell expression of PD-L1 following severe shock attenuates the development of indirect acute lung injury in mice. Am J Physiol Lung Cell Mol Physiol 2020; 318:L801-L812. [PMID: 31994912 PMCID: PMC7191484 DOI: 10.1152/ajplung.00108.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 01/11/2023] Open
Abstract
This study sets out to establish the comparative contribution of PD-L1 expression by pulmonary endothelial cells (ECs) and/or epithelial cells (EpiCs) to the development of indirect acute lung injury (iALI) by taking advantage of the observation that treatment with naked siRNA by intratracheal delivery in mice primarily affects lung EpiCs, but not lung ECs, while intravenous delivery of liposomal-encapsulated siRNA largely targets vascular ECs including the lung, but not pulmonary EpiCs. We showed that using a mouse model of iALI [induced by hemorrhagic shock followed by septic challenge (Hem-CLP)], PD-L1 expression on pulmonary ECs or EpiCs was significantly upregulated in the iALI mice at 24 h post-septic insult. After documenting the selective ability of intratracheal versus intravenous delivery of PD-L1 siRNA to inhibit PD-L1 expression on EpiCs versus ECs, respectively, we observed that the iALI-induced elevation of cytokine/chemokine levels (in the bronchoalveolar lavage fluid, lung lysates, or plasma), lung myeloperoxidase and caspase-3 activities could largely only be inhibited by intravenous, but not intratracheal, delivery of PD-L1 siRNA. Moreover, intravenous, but not intratracheal, delivery led to a preservation of normal tissue architecture, lessened pulmonary edema, and reduced neutrophils influx induced by iALI. In addition, in vitro mouse endothelial cell line studies showed that PD-L1 gene knockdown by siRNA or knockout by CRISPR/Cas9-mediated gene manipulation, reduced monolayer permeability, and maintained tight junction protein levels upon recombinant IFN-γ stimulation. Together, these data imply a critical role for pulmonary vascular ECs in mediating PD-1:PD-L1-driven pathological changes resulting from systemic stimuli such as Hem-CLP.
Collapse
Affiliation(s)
- Shumin Xu
- Department of Emergency Internal Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Yang
- Department of Emergency Internal Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianwen Bai
- Department of Emergency Internal Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianzhu Tao
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
| | - Lunxian Tang
- Department of Emergency Internal Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, the Alpert School of Medicine at Brown University, Providence, Rhode Island
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, the Alpert School of Medicine at Brown University, Providence, Rhode Island
| | - Eleanor A Fallon
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, the Alpert School of Medicine at Brown University, Providence, Rhode Island
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, the Alpert School of Medicine at Brown University, Providence, Rhode Island
| |
Collapse
|
33
|
Abstract
Programed death-ligand 1 (PD-L1, B7-H1, CD274) is a coinhibitory molecule that plays a vital role in the pathogenesis of both neoplastic and nonneoplastic diseases. However, the role of PD-L1 in primary and secondary renal diseases remains to be clarified. Previous studies have shown that both intracellular and intercellular PD-L1 participate in renal diseases via complex mechanisms. PD-L1 plays a dual role in lupus nephritis and has a protective effect in renal ischemia reperfusion injury and nephrotoxic nephritis but not in proliferative immune complex glomerulonephritis. PD-L1 supplementation, anti-PD-L1 antibodies, and D-peptide antagonists have promising application prospects in the treatment of renal diseases. In this review, we summarize the available data published on PD-L1 in renal diseases for the first time.
Collapse
Affiliation(s)
- Yi Wei
- Department of Nephrology, The Sixth affiliated hospital, Sun Yat-sen University, Guangzhou, China
| | - Zongpei Jiang
- Department of Nephrology, The Sixth affiliated hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|