1
|
Jiang H, Guo Y, Wang Q, Wang Y, Peng D, Fang Y, Yan L, Ruan Z, Zhang S, Zhao Y, Zhang W, Shang W, Feng Z. The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions. MedComm (Beijing) 2024; 5:e785. [PMID: 39445002 PMCID: PMC11496570 DOI: 10.1002/mco2.785] [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: 03/31/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
The complement system, comprising over 30 proteins, is integral to the immune system, and the coagulation system is critical for vascular homeostasis. The activation of the complement and coagulation systems involves an organized proteolytic cascade, and the overactivation of these systems is a central pathogenic mechanism in several diseases. This review describes the role of complement and coagulation system activation in critical illness, particularly sepsis. The complexities of sepsis reveal significant knowledge gaps that can be compared to a profound abyss, highlighting the urgent need for further investigation and exploration. It is well recognized that the inflammatory network, coagulation, and complement systems are integral mechanisms through which multiple factors contribute to increased susceptibility to infection and may result in a disordered immune response during septic events in patients. Given the overlapping pathogenic mechanisms in sepsis, immunomodulatory therapies currently under development may be particularly beneficial for patients with sepsis who have concurrent infections. Herein, we present recent findings regarding the molecular relationships between the coagulation and complement pathways in the advancement of sepsis, and propose potential intervention targets related to the crosstalk between coagulation and complement, aiming to provide more valuable treatment of sepsis.
Collapse
Affiliation(s)
- Honghong Jiang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yiming Guo
- Department of Biological Science, The Dietrich School of Arts and SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qihang Wang
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yiran Wang
- Department of Obstetrics and GynecologyThe sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Dingchuan Peng
- School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yigong Fang
- Institute of Acupuncture and MoxibustionChina Academy of Chinese Medical SciencesBeijingChina
| | - Lei Yan
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Zhuolin Ruan
- Department of Obstetrics and Gynecology,Chinese PLA General HospitalBeijingChina
| | - Sheng Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yong Zhao
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Wendan Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Wei Shang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhichun Feng
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| |
Collapse
|
2
|
Yang DJ, Chen KL, Lv ZY, Zhou B, Zhou ZG, Li Y. PD-L1 blockade in mitigating severe acute pancreatitis induced pancreatic damage through modulation of immune cell apoptosis. Int Immunopharmacol 2024; 133:112081. [PMID: 38652963 DOI: 10.1016/j.intimp.2024.112081] [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: 01/04/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Acute pancreatitis (AP) is a prevalent gastrointestinal disorder. The immune response plays a crucial role in AP progression. However, the impact of immune regulatory checkpoint PD-L1 on severe acute pancreatitis (SAP) remains uncertain. Hence, this study aimed to examine the influence of PD-L1 on SAP. We assessed PD-L1 expression in neutrophils and monocytes obtained from SAP patients. We induced SAP in C57BL/6J mice, PD-L1 gene-deficient mice, and PD-L1 humanized mice using intraperitoneal injections of cerulein plus lipopolysaccharide. Prior to the initial cerulein injection, a PD-L1 inhibitor was administered. Pancreatic tissues were collected for morphological and immunohistochemical evaluation, and serum levels of amylase, lipase, and cytokines were measured. Flow cytometry analysis was performed using peripheral blood cells. The expression of PD-L1 in neutrophils and monocytes was significantly higher in SAP patients compared to healthy individuals. Likewise, the expression of PD-L1 in inflammatory cells in the peripheral blood of SAP-induced C57BL/6J mice was notably higher than in the control group. In mice with PD-L1 deficiency, SAP model exhibited lower pancreatic pathology scores, amylase, lipase, and cytokine levels compared to wild-type mice. PD-L1 deletion resulted in reduced neutrophil apoptosis, leading to an earlier peak in neutrophil apoptosis. Furthermore, it decreased early monocyte apoptosis and diminished the peak of T lymphocyte apoptosis. Within the SAP model, administration of a PD-L1 inhibitor reduced pancreatic pathology scores, amylase, lipase, and cytokine levels in both C57BL/6J mice and PD-L1 humanized mice. These findings suggest that inhibiting PD-L1 expression can alleviate the severity of SAP.
Collapse
Affiliation(s)
- Du-Jiang Yang
- Institute of Digestive Surgery, West China Hospital, Sichuan University, No. 1 Ke-yuan-si-lu, Chengdu 610093, Sichuan Province, China; Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, No. 37, Guoxue Alley, Chengdu 610041, Sichuan Province, China
| | - Ke-Ling Chen
- Institute of Digestive Surgery, West China Hospital, Sichuan University, No. 1 Ke-yuan-si-lu, Chengdu 610093, Sichuan Province, China
| | - Zhao-Ying Lv
- Institute of Digestive Surgery, West China Hospital, Sichuan University, No. 1 Ke-yuan-si-lu, Chengdu 610093, Sichuan Province, China
| | - Bin Zhou
- Institute of Digestive Surgery, West China Hospital, Sichuan University, No. 1 Ke-yuan-si-lu, Chengdu 610093, Sichuan Province, China
| | - Zong-Guang Zhou
- Institute of Digestive Surgery, West China Hospital, Sichuan University, No. 1 Ke-yuan-si-lu, Chengdu 610093, Sichuan Province, China
| | - Yuan Li
- Institute of Digestive Surgery, West China Hospital, Sichuan University, No. 1 Ke-yuan-si-lu, Chengdu 610093, Sichuan Province, China.
| |
Collapse
|
3
|
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
|
4
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
5
|
Sun H, Zhang Y, Wang J, Su J, Zhou D, Yu X, Xu Y, Yang W. Application of Lung-Targeted Lipid Nanoparticle-delivered mRNA of soluble PD-L1 via SORT Technology in Acute Respiratory Distress Syndrome. Theranostics 2023; 13:4974-4992. [PMID: 37771784 PMCID: PMC10526659 DOI: 10.7150/thno.86466] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/16/2023] [Indexed: 09/30/2023] Open
Abstract
Rationale: Acute respiratory distress syndrome (ARDS) is a life-threatening condition characterized by excessive immune response usually due to lung inflammation. Local immunosuppression is crucial for effective ARDS treatment. However, current methods are limited in their ability to target the lungs specifically. Methods: This study utilized lung-targeted lipid nanoparticles (LNPs) with 1,2-dioleoyl-3-trimethylammonium-propane (termed DOTAP-LNPs) to encapsulate chemically modified soluble programmed death ligand-1 (sPD-L1) mRNA and examined its physiological characteristics and therapeutic efficacy. A comparative analysis was performed between sPD-L1 mRNA delivered by DOTAP-LNPs, sPD-L1 mRNA delivered by regular LNPs (MC3-LNPs), and PD-L1-Fc recombinant protein administered systemically. Additionally, the survival rate of ARDS mice treated with different drugs was assessed. Results: Administration of sPD-L1 mRNA-LNPs to ARDS model mice significantly reduced leukocyte chemotaxis and protein accumulation in lung tissue, along with a decrease in pulmonary edema. Notably, in situ intervention using sPD-L1 mRNA-DOTAP-LNPs exhibited superior therapeutic effects compared to PD-L1-Fc recombinant protein and sPD-L1 mRNA encapsulated in MC3-LNPs. Importantly, treatment with sPD-L1 mRNA-DOTAP-LNPs improved the survival rate of ARDS model mice. Conclusion: This study demonstrates the feasibility of utilizing stable and reliable mRNA to express the immunosuppressive molecule sPD-L1 specifically in the lungs. The findings provide proof of concept for localized nanoparticle delivery and offer a novel therapeutic strategy for treating acute inflammation in ARDS.
Collapse
Affiliation(s)
- Han Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahui Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Juncheng Su
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Dejian Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Xiang Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai RNACure Biopharma Co., Ltd. Shanghai, 200438, China
- Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases. Shanghai, 200025, China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
6
|
Heffernan DS, Chung CS, Ayala A. SPLENIC INVARIANT NATURAL KILLER T CELLS PLAY A SIGNIFICANT ROLE IN THE RESPONSE TO POLYMICROBIAL SEPSIS. Shock 2023; 60:443-449. [PMID: 37493576 PMCID: PMC10529630 DOI: 10.1097/shk.0000000000002185] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
ABSTRACT Background: Sepsis is marked by a dysregulated immune response to an infection. Invariant natural killer T cells ( i NKT cells) are a pluripotent lymphocyte subpopulation capable of affecting and coordinating the immune response to sepsis. The spleen is an important site of immune interactions in response to an infection. Splenic i NKT cells have emerged as important potential frontline mediators of chronic immune response. There are few data addressing the role splenic of i NKT cells in response to intra-abdominal polymicrobial sepsis. Methods: The cecal ligation and puncture model was used to create intra-abdominal sepsis in 8- to 12-week-old wild-type, i NKT -/- , or programmed cell death receptor-1 (PD-1) -/- mice. Twenty-four hours later, spleens were harvested. Flow cytometry was used for phenotyping using monoclonal antibodies. Cell sort was used to isolate i NKT cells. A macrophage cell line was used to assess i NKT cell-phagocyte interactions. Enzyme-linked immunosorbent assay was used for cytokine analysis. Results: Splenic i NKT-cell populations rapidly declined following induction of sepsis. Within i NKT-cell -/- mice, a distinct baseline hyperinflammatory environment was noted. Within wild type, sepsis induced an increase in splenic IL-6 and IL-10, whereas in i NKT -/- mice, there was no change in elevated IL-6 levels and a noted decrease in IL-10 expression. Further, following sepsis, PD-1 expression was increased upon spleen i NKT cells. With respect to PD-1 ligands upon phagocytes, PD-1 ligand expression was unaffected, whereas PD-L2 expression was significantly affected by the presence of PD-1. Conclusions: Invariant natural killer T cells play a distinct role in the spleen response to sepsis, an effect mediated by the checkpoint protein PD-1. Given that modulators are available in clinical trials, this offers a potential therapeutic target in the setting of sepsis-induced immune dysfunction.
Collapse
Affiliation(s)
- Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Lifespan-Rhode Island Hospital/The Alpert School of Medicine at Brown University, Providence, Rhode Island
| | | | | |
Collapse
|
7
|
Zhong S, Yin Y. Regulatory role of the programmed cell death 1 signaling pathway in sepsis induced immunosuppression. Front Immunol 2023; 14:1183542. [PMID: 37292207 PMCID: PMC10244656 DOI: 10.3389/fimmu.2023.1183542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Sepsis is a multiple organ dysfunction syndrome caused by the host's immune response to infection, with extremely high incidence and mortality. Immunosuppression is an essential pathophysiological alteration that influences the clinical treatment and prognosis of sepsis. Recent studies have suggested that the programmed cell death 1 signaling pathway is involved in the formation of immunosuppression in sepsis. In this review, we systematically present the mechanisms of immune dysregulation in sepsis and elucidate the expression and regulatory effects of the programmed cell death 1 signaling pathway on immune cells associated with sepsis. We then specify current research developments and prospects for the application of the programmed cell death 1 signaling pathway in immunomodulatory therapy for sepsis. Several open questions and future research are discussed at the end.
Collapse
Affiliation(s)
- Shubai Zhong
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuanqin Yin
- Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
8
|
Kim C, Sim H, Bae JS. Benzoylpaeoniflorin Activates Anti-Inflammatory Mechanisms to Mitigate Sepsis in Cell-Culture and Mouse Sepsis Models. Int J Mol Sci 2022; 23:ijms232113130. [PMID: 36361915 PMCID: PMC9656632 DOI: 10.3390/ijms232113130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/29/2022] Open
Abstract
Xuebijing injection (XBJI) (comprising of five herbs) is a widely used traditional Chinese medicine for sepsis treatment. However, the bioactive components of XBJI and the mechanisms responsible for its sepsis-mitigating action have not been experimentally determined. One of the main bioactive compounds in XBJI—benzoylpaeoniflorin (BPF)—inhibits the expressions of key mediators of inflammation such as nuclear factor kappa B (NF-κB), cyclooxygenase-1 (COX-1), and COX-2. However, its effects on sepsis have not been determined yet. Therefore, here, we investigated the immunomodulatory effect of BPF on severely inflamed endothelial cells, THP-1 macrophages, peritoneal macrophages, and mice. Human umbilical vein endothelial cells (HUVECs) and THP-1-macrophages were activated using lipopolysaccharide (LPS) after pretreatment with BPF. Subsequently, changes in the expression profiles of pro-inflammatory molecules including inducible nitric oxide synthase (iNOS), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 were determined using quantitative real-time polymerase chain reaction (qPCR) and Western blot analysis. Furthermore, we monitored the phosphorylation of NF-kB and mitogen-activated protein kinases (MAPKs) to determine their activation levels. Using the LPS-induced mouse model of sepsis, we studied the effects of BPF on inflammatory cytokine production, pulmonary histopathology, and survival rates. Finally, we evaluated whether BPF protects against cecal ligation and puncture (CLP)-induced sepsis, as it closely mimics human sepsis. BPF pretreatment inhibited LPS-induced increase in mRNA and protein levels of iNOS, TNF-α, and IL-6 in HUVECs and THP-1-macrophages. It also suppressed LPS-mediated phosphorylation of p65, p38, JNK, and ERK. Mice with LPS-induced-sepsis who were treated with BPF had lower serum levels of IL-6, TNF-α, IL-1β, CXCL1, and CXCL2 than the control mice treated with BPF. Histopathology revealed that BPF treatment alleviated LPS-induced lung damage. In addition, in mice given a lethal dose of LPS, BPF treatment showed a dose-dependent improvement in survival rates. BPF treatment dose-dependently inhibited the LPS-induced IL-6, TNF-α, and CXCL1 production in peritoneal macrophages. BPF treatment also dose-dependently improved the survival rates in mice with CLP-induced sepsis. These results show that BPF alleviates LPS-stimulated septic conditions and protects mice from CLP-induced sepsis. Our research marks BPF as a potential drug in the treatment of sepsis and various inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Jong-Sup Bae
- Correspondence: ; Tel.: +82-53-950-8570; Fax: +82-53-950-8557
| |
Collapse
|
9
|
Zhang P, Wang Y, Yang W, Yin Y, Li C, Ma X, Shi L, Li R, Tao K. 4-Octyl itaconate regulates immune balance by activating Nrf2 and negatively regulating PD-L1 in a mouse model of sepsis. Int J Biol Sci 2022; 18:6189-6209. [PMID: 36439878 PMCID: PMC9682535 DOI: 10.7150/ijbs.74456] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022] Open
Abstract
Introduction: Sepsis is a major global health challenge with high mortality rates and no effective treatment. Recent studies have suggested that sepsis may be associated with immune system dysfunction. Itaconate may exert anti-inflammatory effects via Nrf2 signaling. Although Nrf2 regulates oxidative/exogenous stress responses and inhibits inflammatory responses, the mechanism via which Nrf2 regulates immune checkpoints in sepsis remains unclear. Objectives: This study aimed to investigate the role of the Nrf2 signaling pathway in sepsis immunosuppression injury by exploring Nrf2 target genes in inflammatory macrophages in a mouse model of sepsis. Methods: We evaluated the effects of 4-octyl itaconate (OI) on pro-inflammatory and anti-inflammatory cytokines in a mouse model of sepsis and RAW264.7 cells. In addition, we investigated if OI could inhibit LPS-induced oxidative stress by activating Nrf2 signaling in vitro and in vivo. Results: OI reduced the release of pro-inflammatory cytokines and increased the release of anti-inflammatory cytokines, thereby inhibiting inflammation. OI increased glutathione synthase (GSS) expression by activating the Nrf2 signaling pathway to promote GSH synthesis, thus, inhibiting oxidative stress. OI inhibited the early release of inflammatory and oxidative stress-related factors to reduce tissue and organ injury in mice with sepsis, while Nrf2 interfered with PD-L1 induction and inhibited PD-L1 expression at an advanced stage to reduce the occurrence of sepsis immunosuppression. Conclusions: This study indicates that Nrf2 is a novel negative regulator of PD-L1 that functions at immune checkpoints and suggests an underlying mechanism for the anti-inflammatory process mediated by Nrf2.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wengchang Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chengguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xianxiong Ma
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
10
|
Ou H, Chen K, Chen L, Wu H. Bioinformatic analysis of PD-1 checkpoint blockade response in influenza infection. BMC Genom Data 2022; 23:65. [PMID: 35962325 PMCID: PMC9374577 DOI: 10.1186/s12863-022-01081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 06/22/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The programmed cell death 1 (PD-1)/PD-1 ligand 1 (PD-L1) signaling pathway is significantly upregulated in influenza virus infection, which impairs the antiviral response. Blocking this signaling pathway may reduce the damage, lower the virus titer in lung tissue, and alleviate the symptoms of infection to promote recovery. In addition to the enhanced viral immune response, using of immune checkpoint inhibitors in influenza virus infection is controversial, the aim of this study was to identify the key factors and regulatory mechanisms in the PD-1 checkpoint blockade response microenvironment in influenza infection. METHODS A BALB/c mouse model of influenza A/PR8(H1N1) infection was established then constructed, and whole-transcriptome sequencing including mRNAs, miRNAs (microRNAs), lncRNAs (long noncoding RNAs), and circRNAs (circular RNAs) of mice treated with PD-1 checkpoint blockade by antibody treatment and IgG2a isotype control before infection with A/PR8(H1N1) were performed. Subsequently, the differential expression of transcripts between these two groups was analyzed, followed by functional interaction prediction analysis to investigate gene-regulatory circuits. RESULTS In total, 84 differentially expressed dif-mRNAs, 36 dif-miRNAs, 90 dif-lncRNAs and 22 dif-circRNAs were found in PD-1 antagonist treated A/PR8(H1N1) influenza-infected lungs compared with the controls (IgG2a isotype control treated before infection). In spleens between the above two groups, 45 dif-mRNAs, 36 dif-miRNAs, 57 dif-lncRNAs, and 24 dif-circRNAs were identified. Direct function enrichment analysis of dif-mRNAs and dif-miRNAs showed that these genes were mainly involved in myocardial damage related to viral infection, mitogen activated protein kinase (MAPK) signaling pathways, RAP1 (Ras-related protein 1) signaling pathway, and Axon guidance. Finally, 595 interaction pairs were obtained for the lungs and 462 interaction pairs for the spleens were obtained in the competing endogenous RNA (ceRNA) complex network, in which the downregulated mmu-miR-7043-3p and Vps39-204 were enriched significantly in PD-1 checkpoint blockade treated A/PR8(H1N1) infection group. CONCLUSIONS The present study provided a basis for the identification of potential pathways and hub genes that might be involved in the PD-1 checkpoint blockade response microenvironment in influenza infection.
Collapse
Affiliation(s)
- Huilin Ou
- Ningbo Medical Centre, Li Huili Hospital affiliated of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Linfang Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310015, China
| | - Hongcheng Wu
- Ningbo Medical Centre, Li Huili Hospital affiliated of Ningbo University, Ningbo, 315040, Zhejiang, China.
| |
Collapse
|
11
|
Ahn JH, Song EJ, Jung DH, Kim YJ, Seo IS, Park SC, Jung YS, Cho ES, Mo SH, Hong JJ, Cho JY, Park JH. The sesquiterpene lactone estafiatin exerts anti-inflammatory effects on macrophages and protects mice from sepsis induced by LPS and cecal ligation puncture. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153934. [PMID: 35172258 DOI: 10.1016/j.phymed.2022.153934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Previously, we found that the water extract of Artermisia scoparia Waldst. & Kit suppressed the cytokine production of lipopolysaccharide (LPS)-stimulated macrophages and alleviated carrageenan-induced acute inflammation in mice. Artemisia contains various sesquiterpene lactones and most of them exert immunomodulatory activity. PURPOSE In the present study, we investigated the immunomodulatory effect of estafiatin (EST), a sesquiterpene lactone derived from A. scoparia, on LPS-induced inflammation in macrophages and mouse sepsis model. STUDY DESIGN AND METHODS Murine bone marrow-derived macrophages (BMDMs) and THP-1 cells, a human monocytic leukemia cell line, were pretreated with different doses of EST for 2 h, followed by LPS treatment. The gene and protein expression of pro-inflammatory cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α, and inducible nitric oxide synthase (iNOS) were measured by quantitative real-time polymerase chain reaction (qPCR) and Western blot analysis. The activation of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) was also evaluated at the level of phosphorylation. The effect of EST on inflammatory cytokine production, lung histopathology, and survival rate was assessed in an LPS-induced mice model of septic shock. The effect of EST on the production of cytokines in LPS-stimulated peritoneal macrophages was evaluated by in vitro and ex vivo experiments and protective effect of EST on cecal ligation and puncture (CLP) mice was also assessed. RESULTS The LPS-induced expression of IL-6, TNF-α, and iNOS was suppressed at the mRNA and protein levels in BMDMs and THP-1 cells, respectively, by pretreatment with EST. The half-maximal inhibitory concentration (IC50) of EST on IL-6 and TNF-α production were determined as 3.2 μM and 3.1 μM in BMDMs, 3 μM and 3.4 μM in THP1 cells, respectively. In addition, pretreatment with EST significantly reduced the LPS-induced phosphorylation p65, p38, JNK, and ERK in both cell types. In the LPS-induced mice model of septic shock, serum levels of IL-6, TNF-α, IL-1β, CXCL1, and CXCL2 were lower in EST-treated mice than in the control animals. Histopathology analysis revealed that EST treatment ameliorated LPS-induced lung damage. Moreover, while 1 of 7 control mice given lethal dose of LPS survived, 3 of 7 EST-treated (1.25 mg/kg) mice and 5 of 7 EST-treated (2.5 mg/kg) mice were survived. Pretreatment of EST dose-dependently suppressed the LPS-induced production of IL-6, TNF-α and CXCL1 in peritoneal macrophages. In CLP-induced mice sepsis model, while all 6 control mice was dead at 48 h, 1 of 6 EST-treated (1.25 mg/kg) mice and 3 of 6 EST-treated (2.5 mg/kg) mice survived for 96 h. CONCLUSION These results demonstrated that EST exerts anti-inflammatory effects on LPS-stimulated macrophages and protects mice from sepsis. Our study suggests that EST could be developed as a new therapeutic agent for sepsis and various inflammatory diseases.
Collapse
Affiliation(s)
- Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Eun-Jung Song
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Yeong-Jun Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - In-Su Seo
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Seong-Chan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - You-Seok Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Eun-Seo Cho
- Department of Food Science & Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Sang Hyun Mo
- Department of Food Science & Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk 28116, Republic of Korea.
| | - Jeong-Yong Cho
- Department of Food Science & Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
12
|
Thylur Puttalingaiah R. Role of Swiprosin-1/EFHD2 as a biomarker in the development of chronic diseases. Life Sci 2022; 297:120462. [PMID: 35276221 DOI: 10.1016/j.lfs.2022.120462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
Swiprosin-1 or EFHD2, is a Ca2+ binding actin protein and its expression has been shown to be distinct in various cell types. The expression of swiprosin-1 is upregulated during the activation of immune cells, epithelial and endothelial cells. The expression of swiprosin-1 is regulated by diverse signaling pathways that are contingent upon the specific type of cells. The aim of this review is to summarize and provide an overview of the role of swiprosin-1 in pathophysiological conditions of cancers, cardiovascular diseases, diabetic nephropathy, neuropsychiatric diseases, and in the process of inflammation, immune response, and inflammatory diseases. Novel approaches for the targeting of swiprosin-1 as a biomarker in the early detection and prevention of various development of chronic diseases are also explored.
Collapse
Affiliation(s)
- Ramesh Thylur Puttalingaiah
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, 1700 Tulane Avenue, Room 945-B1, New Orleans, LA 70112, USA..
| |
Collapse
|
13
|
Tilstam PV, Schulte W, Holowka T, Kim BS, Nouws J, Sauler M, Piecychna M, Pantouris G, Lolis E, Leng L, Bernhagen J, Fingerle-Rowson G, Bucala R. MIF but not MIF-2 recruits inflammatory macrophages in an experimental polymicrobial sepsis model. J Clin Invest 2021; 131:127171. [PMID: 34850744 DOI: 10.1172/jci127171] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
Excessive inflammation drives the progression from sepsis to septic shock. Macrophage migration inhibitory factor (MIF) is of interest because MIF promoter polymorphisms predict mortality in different infections, and anti-MIF antibody improves survival in experimental models when administered 8 hours after infectious insult. The recent description of a second MIF superfamily member, D-dopachrome tautomerase (D-DT/MIF-2), prompted closer investigation of MIF-dependent responses. We subjected Mif-/- and Mif-2-/- mice to polymicrobial sepsis and observed a survival benefit with Mif but not Mif-2 deficiency. Survival was associated with reduced numbers of small peritoneal macrophages (SPMs) that, in contrast to large peritoneal macrophages (LPMs), were recruited into the peritoneal cavity. LPMs produced higher quantities of MIF than SPMs, but SPMs expressed higher levels of inflammatory cytokines and the MIF receptors CD74 and CXCR2. Adoptive transfer of WT SPMs into Mif-/- hosts reduced the protective effect of Mif deficiency in polymicrobial sepsis. Notably, MIF-2 lacks the pseudo-(E)LR motif present in MIF that mediates CXCR2 engagement and SPM migration, supporting a specific role for MIF in the recruitment and accumulation of inflammatory SPMs.
Collapse
Affiliation(s)
- Pathricia Veronica Tilstam
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Wibke Schulte
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Charité, Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Thomas Holowka
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bong-Sung Kim
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Plastic, Reconstructive and Hand Surgery, RWTH Aachen University, Aachen, Germany.,Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jessica Nouws
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maor Sauler
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marta Piecychna
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Georgios Pantouris
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Chemistry, University of the Pacific, Stockton, California, USA
| | - Elias Lolis
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| | - Günter Fingerle-Rowson
- Department I of Internal Medicine, University of Cologne, Center for Integrated Oncology Aachen Bonn Köln Düsseldorf, Cologne, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Dellinger RP, Levy MM, Schorr CA, Townsend SR. 50 Years of Sepsis Investigation/Enlightenment Among Adults-The Long and Winding Road. Crit Care Med 2021; 49:1606-1625. [PMID: 34342304 DOI: 10.1097/ccm.0000000000005203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- R Phillip Dellinger
- Cooper Medical School of Rowan University and Cooper University Health, Camden, NJ
| | | | - Christa A Schorr
- Cooper Medical School of Rowan University and Cooper University Health, Camden, NJ
| | - Sean R Townsend
- University of California Pacific Medical Center, (Sutter Health), San Francisco, CA
| |
Collapse
|
15
|
Chen R, Zhou L. PD-1 signaling pathway in sepsis: Does it have a future? Clin Immunol 2021; 229:108742. [PMID: 33905818 DOI: 10.1016/j.clim.2021.108742] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/07/2021] [Accepted: 04/22/2021] [Indexed: 01/17/2023]
Abstract
Sepsis is characterized by high mortality and poor prognosis and is one of the leading causes of death among patients in the intensive care unit (ICU). In the past, drugs that block early inflammatory responses have done little to reverse the progression of sepsis. Programmed cell death receptor 1 (PD-1) and its two ligands, programmed cell death receptor ligand 1(PD-L1) and programmed cell death receptor ligand 2 (PD-L2), are negative regulatory factors of the immune response of the body. Recently, the role of the PD-1 signaling pathway in sepsis has been widely studied. Studies showed that the PD-1 signaling pathways are closely related to the mortality and prognosis of sepsis patients. In the immunotherapy of sepsis, whether in animal experiments or clinical trials, anti-PD-1/PD-L1 antibodies have shown good promise. In this review, firstly, we focus on the immunosuppressive mechanism of sepsis and the structure and function of the PD-1 signaling pathway. The variety of the PD-1 signaling pathways in sepsis is introduced. Then, the relationship between the PD-1 signaling pathway and immune cells and organ dysfunction and the regulatory factors of the PD-1 signaling pathway in sepsis is discussed. Finally, the application of the PD-1 signaling pathway in sepsis is specifically emphasized.
Collapse
Affiliation(s)
- Rongping Chen
- Department of Intensive care unit, The First People's Hospital of Foshan, Foshan 528000, Guangdong Province, China; Sun Yet-sen University, Guangzhou 510000, Guangdong Province, China
| | - Lixin Zhou
- Department of Intensive care unit, The First People's Hospital of Foshan, Foshan 528000, Guangdong Province, China.
| |
Collapse
|
16
|
Abstract
Sepsis is a life-threatening syndrome with a high incidence and a weighty economic burden. The cytokines storm in the early stage and the state of immunosuppression in the late stage contribute to the mortality of sepsis. Immune checkpoints expressed on lymphocytes and APCs, including CD28, CTLA-4, CD80, CD86, PD-1 and PD-L1, CD40 and CD40L, OX40 and OX40L, 4-1BB and 4-1BBL, BTLA, TIM family, play significant roles in the pathogenesis of sepsis through regulating the immune disorder. The specific therapies targeting immune checkpoints exhibit great potentials in the animal and preclinical studies, and further clinical trials are planning to implement. Here, we review the current literature on the roles played by immune checkpoints in the pathogenesis and treatment of sepsis. We hope to provide further insights into this novel immunomodulatory strategy.
Collapse
Affiliation(s)
- Yan-Cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Song-Tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-Fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
17
|
Xu J, Wang J, Wang X, Tan R, Qi X, Liu Z, Qu H, Pan T, Zhan Q, Zuo Y, Yang W, Liu J. Soluble PD-L1 improved direct ARDS by reducing monocyte-derived macrophages. Cell Death Dis 2020; 11:934. [PMID: 33127884 PMCID: PMC7596316 DOI: 10.1038/s41419-020-03139-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/02/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is common in intensive care units (ICUs), although it is associated with high mortality, no effective pharmacological treatments are currently available. Despite being poorly understood, the role of programmed cell death protein 1 (PD-1) and PD-ligand 1 (PD-L1) axis in ARDS may provide significant insights into the immunosuppressive mechanisms that occur after ARDS. In the present study, we observed that the level of soluble PD-L1 (sPD-L1), a potential activator of the PD-1 pathway, was upregulated in survivors of direct ARDS than in non-survivors. Administration of sPD-L1 in mice with direct ARDS relieved inflammatory lung injury and improved the survival rate, indicating the protective role of sPD-L1 in direct ARDS. Using high-throughput mass cytometry, we found a marked decrease in the number of lung monocyte-derived macrophages (MDMs) with proinflammatory markers, and the protective role of sPD-L1 diminished in ARDS mice with monocyte/macrophage depletion. Furthermore, PD-1 expression increased in the MDMs of patients and mice with direct ARDS. Finally, we showed that sPD-L1 induced MDM apoptosis in patients with direct ARDS. Taken together, our results demonstrated that the engagement of sPD-L1 on PD-1 expressing macrophages resulted in a decrease in pro-inflammatory macrophages and eventually improved direct ARDS. Our study identified a prognostic indicator for patients with direct ARDS and a potential target for therapeutic development in direct ARDS.
Collapse
Affiliation(s)
- Jing Xu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahui Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoling Qi
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wen Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jialin Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Zhang Q, Sun H, Zhuang S, Liu N, Bao X, Liu X, Ren H, Lv D, Li Z, Bai J, Zhou X, Tang L. Novel pharmacological inhibition of EZH2 attenuates septic shock by altering innate inflammatory responses to sepsis. Int Immunopharmacol 2019; 76:105899. [PMID: 31518916 DOI: 10.1016/j.intimp.2019.105899] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/06/2019] [Accepted: 09/06/2019] [Indexed: 01/11/2023]
Abstract
The function of histone methyltransferase enhancer of zeste homolog 2 (EZH2) in sepsis remains unknown. We reported here that the expression of EZH2 and H3K27me3 was significantly upregulated in the circulation of septic patients, whereas patients who survived presented downregulated the expression of EZH2 on CD14+ monocytes. We further identified increased expression of EZH2 in the circulation, peritoneal fluid, and septic lungs from CLP mice. 3-DZNeP treated CLP mice improved mortality and protected from organ injury. EZH2 inhibition not only suppressed the activation of inflammatory cells and release of cytokines in the circulation and infectious sites, but also promoted bacteria clearance and replenished the circulating monocyte and neutrophil pool from bone marrow. Blockage of EZH2 also suppressed the progression of lung injury and alleviated inflammation by decreasing the pulmonary cell apoptosis, reducing inflammatory cells infiltration and cytokines release through inhibition of the STAT3 signaling pathway and recovery of PPARγ activation. In addition, EZH2 inhibitor blunted macrophage M1 polarization by SOCS3/STAT1 pathway. Overall, these data suggest that EZH2 could be a potential biomarker predicting clinical outcome and a new target for therapeutic interference in sepsis.
Collapse
Affiliation(s)
- Qianqian Zhang
- Medical School/Tongji University, Shanghai 200120, China
| | - Hong Sun
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China; Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaowei Bao
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China
| | - Xiandong Liu
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China
| | - Huijuan Ren
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China
| | - Diyu Lv
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China
| | - Zhe Li
- Medical School/Tongji University, Shanghai 200120, China
| | - Jianwen Bai
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University, Shanghai 200120, China.
| | - Lunxian Tang
- Department of Internal Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai 200120, China; Department of Internal Emergency Medicine and Critical Care, ji'an Hospital, Shanghai East Hospital, ji'an, jiangxi, China.
| |
Collapse
|
19
|
Fallon EA, Biron-Girard BM, Chung CS, Lomas-Neira J, Heffernan DS, Monaghan SF, Ayala A. A novel role for coinhibitory receptors/checkpoint proteins in the immunopathology of sepsis. J Leukoc Biol 2018; 103:10.1002/JLB.2MIR0917-377R. [PMID: 29393983 PMCID: PMC6314914 DOI: 10.1002/jlb.2mir0917-377r] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/26/2017] [Accepted: 01/03/2018] [Indexed: 12/11/2022] Open
Abstract
Coinhibitory molecules, such as PD-1, CTLA-4, 2B4, and BTLA, are an important new family of mediators in the pathophysiology of severe bacterial and/or fungal infection, as well as the combined insults of shock and sepsis. Further, the expression of these molecules may serve as indicators of the immune status of the septic individual. Using PD-1:PD-L as an example, we discuss in this review how such checkpoint molecules may affect the host response to infection by mediating the balance between effective immune defense and immune-mediated tissue injury. Additionally, we explore how the up-regulation of PD-1 and/or PD-L1 expression on not only adaptive immune cells (e.g., T cells), but also on innate immune cells (e.g., macrophages, monocytes, and neutrophils), as well as nonimmune cells during sepsis and/or shock contributes to functional alterations often with detrimental sequelae.
Collapse
Affiliation(s)
- Eleanor A. Fallon
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| | - Bethany M. Biron-Girard
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| | - Joanne Lomas-Neira
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| | - Daithi S. Heffernan
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| | - Sean F. Monaghan
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Providence, R.I., USA
| |
Collapse
|
20
|
Zheng YH, Deng YY, Lai W, Zheng SY, Bian HN, Liu ZA, Huang ZF, Sun CW, Li HH, Luo HM, Ma LH, Chen HX, Xiong B. Effect of bone marrow mesenchymal stem cells on the polarization of macrophages. Mol Med Rep 2018; 17:4449-4459. [PMID: 29363724 PMCID: PMC5802220 DOI: 10.3892/mmr.2018.8457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 12/01/2017] [Indexed: 12/23/2022] Open
Abstract
Inflammation is a defensive response in the living tissue of the vascular system that acts against damage factors and involves various types of immune cells, including macrophages, neutrophils, endothelial cells and other associated immune molecules. If the release of inflammatory mediators is excessive, systemic inflammatory response syndrome may develop. Sepsis is the most common complication of severe burns and is a systemic inflammatory response syndrome that is caused by infectious factors and is capable of leading to multiple organ dysfunction and potentially death. Research concerning the mechanism and treatment of sepsis is crucial. Macrophages are an important type of immune cell that remove invasive pathogens and are involved in innate and adaptive immune responses. It has been previously reported that bone marrow mesenchymal stem cells (BMSCs) affect macrophages by regulating immunity. The present study aimed to investigate the effect of BMSCs on macrophage polarization in vivo and in vitro, in addition to the potential therapeutic effect of these cells on experimental sepsis. BMSCs and peritoneal macrophages were isolated from Sprague‑Dawley rats and co‑cultured overnight as a mixed culture or Transwell system, and subsequently stimulated with 100 ng/ml lipopolysaccharide (LPS). After 12 h, the medium was replaced with normal complete medium for various durations and supernatants were collected to extract proteins and cells for ELISA, western blot and flow cytometry analysis to investigate different aspects of macrophages. Sepsis was induced in Sprague‑Dawley rats by injection of LPS (5 mg/kg), followed by tail vein injection of BMSCs or PBS 1 h later. After 6, 12, 24 and 48 h, lung tissues were harvested for pathological observation and peritoneal macrophages were collected for flow cytometry analysis to assess the expression of markers, including cluster of differentiation (CD)68 (used for gating), CD11c and CD206. The results demonstrated that, in the culture medium, LPS stimulation increased the expression of CD11c in macrophages, and the levels of tumor necrosis factor‑α and inducible nitric oxide synthase were also increased. By contrast, in macrophages treated with BMSCs directly, the expression of CD11c was reduced compared with the LPS‑stimulated macrophage alone group. However, the secretion of interleukin‑10, transforming growth factor‑β and arginase‑1 was increased in the direct co‑culture group, compared with the LPS‑stimulated macrophage alone group. BMSCs reduced the inflammation in lung tissues and inhibited macrophage expression of CD11c in the rat model of sepsis. The results of the present study demonstrated that BMSCs co‑cultured with macrophages directly inhibited macrophage differentiation into the M1 phenotype and reduced inflammation in macrophages stimulated by LPS. In vivo, BMSCs decreased the expression of CD11c in peritoneal macrophages and reduced the pathological inflammatory response in the lungs. The findings of the present study demonstrated that BMSCs may reduce the extent of the systemic inflammatory response, which may contribute to the development for a novel type of treatment for sepsis in the future.
Collapse
Affiliation(s)
- Yuan Hua Zheng
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yi Yu Deng
- Department of Critical Care and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Wen Lai
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Shao Yi Zheng
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Hui Ning Bian
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zu An Liu
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhi Feng Huang
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Chuan Wei Sun
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Han Hua Li
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Hong Min Luo
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Liang Hua Ma
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Han Xi Chen
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Bing Xiong
- Department of Burns and Wound Repair Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
21
|
MiR-142 inhibits cecal ligation and puncture (CLP)-induced inflammation via inhibiting PD-L1 expression in macrophages and improves survival in septic mice. Biomed Pharmacother 2018; 97:1479-1485. [DOI: 10.1016/j.biopha.2017.11.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/27/2017] [Accepted: 11/10/2017] [Indexed: 12/11/2022] Open
|
22
|
Tu Y, Zhang L, Tong L, Wang Y, Zhang S, Wang R, Li L, Wang Z. EFhd2/swiprosin-1 regulates LPS-induced macrophage recruitment via enhancing actin polymerization and cell migration. Int Immunopharmacol 2017; 55:263-271. [PMID: 29288926 DOI: 10.1016/j.intimp.2017.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 12/25/2022]
Abstract
Macrophage motility is vital in innate immunity, which contributes strategically to the defensive inflammation process. During bacterial infection, lipopolysaccharide (LPS) potently activates the migration of macrophages via the NF-κB/iNOS/c-Src signaling pathway. However, the downstream region of c-Src that participates in macrophage migration is unclear. EFhd2, a novel actin bundling protein, was evaluated for its role in LPS-stimulated macrophage migration in this study. We found that LPS stimulated the up-regulation, tyrosine phosphorylation and membrane translocation of EFhd2 in macrophages. The absence of EFhd2 inhibited the recruitment of macrophages in the lungs of LPS-induced septic mice. LPS-induced macrophage migration was neutralized by the deletion of EFhd2. EFhd2-mediated up-regulation of NFPs (including Rac1/Cdc42, N-WASP/WAVE2 and Arp2/3 complex) induced by LPS could be used to explain the role of EFhd2 in promoting actin polymerization. Furthermore, the purified EFhd2 could directly promote actin polymerization in vitro. Dasatinib, a c-Src specific inhibitor, inhibited the up-regulation of EFhd2 stimulated by LPS. Therefore, our study demonstrated that EFhd2 might be involved in LPS-stimulated macrophage migration, which provides a potential target for LPS-activated c-Src during macrophage mobilization.
Collapse
Affiliation(s)
- Ye Tu
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China; Department of Medical Department, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Lichao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Lingchang Tong
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yue Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Su Zhang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Rongmei Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ling Li
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Zhibin Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
23
|
Young WA, Fallon EA, Heffernan DS, Efron PA, Cioffi WG, Ayala A. Improved survival after induction of sepsis by cecal slurry in PD-1 knockout murine neonates. Surgery 2016; 161:1387-1393. [PMID: 28012568 DOI: 10.1016/j.surg.2016.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/12/2016] [Accepted: 11/05/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Sepsis and the ensuing immune dysfunction continue to be major contributors to neonatal morbidity and mortality. Neonatal sepsis also is associated with profound immune dysfunction. We have recently identified a role for a family of coinhibitory molecules that are altered in murine sepsis and in critically ill adult patients, which may be a target for development of novel therapies. There is, however, a paucity of data pertaining to the role of coinhibitory checkpoint proteins in the control and modulation of neonatal sepsis. METHODS The cecal slurry model consists of harvesting the cecal content of an adult, wild-type, male mouse and combining it with 5% dextrose to create a cecal slurry with a concentration of 80 mg/mL (LD70 at 7 days). Neonatal mice (5-7 days of age) underwent intraperitoneal injection of the cecal slurry or 0.9% saline for the sham procedure. Wild-type (C57BL/6) or PD-1-/- mice were used; a 7-day survival study was undertaken. Cytometric bead array was used for cytokine expression. Blood and peritoneal fluid was cultured for bacterial burden. Flow cytometry was used to assess the peritoneal cavity cell populations. RESULTS There was no mortality after the sham procedure in either wild-type or PD-1-/- pups. PD-1 markedly affected sepsis survival with significantly improved survival in the PD-1-/- pups (40% vs 80%; P < .01). This survival improvement was not associated with any difference in bacterial clearance. The bacterial burden was equivalent between wild-type and PD-1-/- pups at 24 hours after cecal slurry. However, PD-1-/- pups did display an increased circulating cytokine response to the cecal slurry compared with wild type, with increased expression of IL-6, IL-10, and TNF-α levels. Within the peritoneal cavity, sepsis induced an influx of neutrophils, a finding that was increased in PD-1-/- pups. Although the T-cell response was unaffected by PD-1, it was noted that cecal slurry induced a loss of peritoneal B cells in WT, while the peritoneal B-cell population was preserved in PD-1-/- pups. CONCLUSION Our data suggest that the checkpoint protein, PD-1, plays an important role in controlling the immune response to sepsis in the neonate, ultimately affecting sepsis-related mortality in this neonatal murine model of sepsis. Akin to adult studies, these data further emphasize the potential therapeutic target for PD-1 across a spectrum of septic patients.
Collapse
Affiliation(s)
- Whitney A Young
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI
| | - Eleanor A Fallon
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI
| | - Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI
| | - Philip A Efron
- Laboratory of Inflammation Biology and Surgical Science, Departments of Surgery, Anesthesia, Aging, and Geriatric Research, and Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL
| | - William G Cioffi
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Brown University, Rhode Island Hospital, Providence, RI.
| |
Collapse
|
24
|
Wang F, Huang X, Chung CS, Chen Y, Hutchins NA, Ayala A. Contribution of programmed cell death receptor (PD)-1 to Kupffer cell dysfunction in murine polymicrobial sepsis. Am J Physiol Gastrointest Liver Physiol 2016; 311:G237-45. [PMID: 27288425 PMCID: PMC5007287 DOI: 10.1152/ajpgi.00371.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 06/07/2016] [Indexed: 01/31/2023]
Abstract
Recent studies suggest that coinhibitory receptors appear to be important in contributing sepsis-induced immunosuppression. Our laboratory reported that mice deficient in programmed cell death receptor (PD)-1 have increased bacterial clearance and improved survival in experimental sepsis induced by cecal ligation and puncture (CLP). In response to infection, the liver clears the blood of bacteria and produces cytokines. Kupffer cells, the resident macrophages in the liver, are strategically situated to perform the above functions. However, it is not known if PD-1 expression on Kupffer cells is altered by septic stimuli, let alone if PD-1 ligation contributes to the altered microbial handling seen. Here we report that PD-1 is significantly upregulated on Kupffer cells during sepsis. PD-1-deficient septic mouse Kupffer cells displayed markedly enhanced phagocytosis and restoration of the expression of major histocompatibility complex II and CD86, but reduced CD80 expression compared with septic wild-type (WT) mouse Kupffer cells. In response to ex vivo LPS stimulation, the cytokine productive capacity of Kupffer cells derived from PD-1-/- CLP mice exhibited a marked, albeit partial, restoration of the release of IL-6, IL-12, IL-1β, monocyte chemoattractant protein-1, and IL-10 compared with septic WT mouse Kupffer cells. In addition, PD-1 gene deficiency decreased LPS-induced apoptosis of septic Kupffer cells, as indicated by decreased levels of cleaved caspase-3 and reduced terminal deoxynucleotidyl transferase dUTP nick end-labeling-positive cells. Exploring the signal pathways involved, we found that, after ex vivo LPS stimulation, septic PD-1-/- mouse Kupffer cells exhibited an increased Akt phosphorylation and a reduced p38 phosphorylation compared with septic WT mouse Kupffer cells. Together, these results indicate that PD-1 appears to play an important role in regulating the development of Kupffer cell dysfunction seen in sepsis.
Collapse
Affiliation(s)
- Fei Wang
- 1Department of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, PR China; and
| | - Xin Huang
- 2Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, Rhode Island
| | - Chun-Shiang Chung
- 2Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, Rhode Island
| | - Yaping Chen
- 2Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, Rhode Island
| | - Noelle A. Hutchins
- 2Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, Rhode Island
| | - Alfred Ayala
- Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
25
|
Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A. The new normal: immunomodulatory agents against sepsis immune suppression. Trends Mol Med 2014; 20:224-33. [PMID: 24485901 DOI: 10.1016/j.molmed.2014.01.002] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 12/30/2013] [Accepted: 01/03/2014] [Indexed: 12/15/2022]
Abstract
Sepsis is the leading cause of death among critically ill patients in intensive care units, and treatment options are limited. Therapies developed against the proinflammatory stage have failed clinically; therefore, new approaches that target the host immune response in sepsis are necessary. Increasing evidence suggests that a major pathophysiological event in sepsis is immune suppression, often resulting in secondary fungal, bacterial, or viral infections. Recent studies from animal sepsis models and patient samples suggest that cytokines such as interleukin-7 (IL-7), IL-15, granulocyte macrophage colony-stimulating factor (GM-CSF), as well as co-inhibitory molecule blockade, such as anti-programmed cell death receptor-1 (anti-PD-1) and anti-B and T lymphocyte attenuator (anti-BTLA), may have utility in alleviating the clinical morbidity associated with sustained sepsis. This review discusses some of these novel immunomodulatory agents and evaluates their potential use as therapeutics.
Collapse
Affiliation(s)
- Noelle A Hutchins
- Division of Surgical Research, Rhode Island Hospital, Providence, RI 02903, USA
| | - Jacqueline Unsinger
- Department of Anesthesiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Richard S Hotchkiss
- Department of Anesthesiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Alfred Ayala
- Division of Surgical Research, Rhode Island Hospital, Providence, RI 02903, USA.
| |
Collapse
|