1
|
Kang S, Onishi S, Ling Z, Inoue H, Zhang Y, Chang H, Zhao H, Wang T, Okuzaki D, Matsuura H, Takamatsu H, Oda J, Kishimoto T. Gp130-HIF1α axis-induced vascular damage is prevented by the short-term inhibition of IL-6 receptor signaling. Proc Natl Acad Sci U S A 2024; 121:e2315898120. [PMID: 38165930 PMCID: PMC10786312 DOI: 10.1073/pnas.2315898120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/22/2023] [Indexed: 01/04/2024] Open
Abstract
Protection against endothelial damage is recognized as a frontline approach to preventing the progression of cytokine release syndrome (CRS). Accumulating evidence has demonstrated that interleukin-6 (IL-6) promotes vascular endothelial damage during CRS, although the molecular mechanisms remain to be fully elucidated. Targeting IL-6 receptor signaling delays CRS progression; however, current options are limited by persistent inhibition of the immune system. Here, we show that endothelial IL-6 trans-signaling promoted vascular damage and inflammatory responses via hypoxia-inducible factor-1α (HIF1α)-induced glycolysis. Using pharmacological inhibitors targeting HIF1α activity or mice with the genetic ablation of gp130 in the endothelium, we found that inhibition of IL-6R (IL-6 receptor)-HIF1α signaling in endothelial cells protected against vascular injury caused by septic damage and provided survival benefit in a mouse model of sepsis. In addition, we developed a short half-life anti-IL-6R antibody (silent anti-IL-6R antibody) and found that it was highly effective at augmenting survival for sepsis and severe burn by strengthening the endothelial glycocalyx and reducing cytokine storm, and vascular leakage. Together, our data advance the role of endothelial IL-6 trans-signaling in the progression of CRS and indicate a potential therapeutic approach for burns and sepsis.
Collapse
Affiliation(s)
- Sujin Kang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
- Department of Immune Regulation, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka565-0871, Japan
| | - Shinya Onishi
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka565-0871, Japan
| | - Zhenzhen Ling
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Hitomi Inoue
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Yingying Zhang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Hao Chang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Hui Zhao
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Tong Wang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Daisuke Okuzaki
- Next Generation-Sequencing Core Facility, Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
| | - Hiroshi Matsuura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka565-0871, Japan
| | - Hyota Takamatsu
- Department of Clinical Research Center for Autoimmune Disease, Osaka Minami Medical Center, National Hospital Organization, Kawachinagano, Osaka586-8521, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Jun Oda
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka565-0871, Japan
| | - Tadamitsu Kishimoto
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
- Department of Immune Regulation, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka565-0871, Japan
| |
Collapse
|
2
|
Liu X, Liu Y, Li X, Huang J, Guo X, Zhang J, Luo Z, Shi Y, Jiang M, Qin B, Du Y, Luo L, You J. ER-Targeting PDT Converts Tumors into In Situ Therapeutic Tumor Vaccines. ACS NANO 2022; 16:9240-9253. [PMID: 35713245 DOI: 10.1021/acsnano.2c01669] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A therapeutic tumor vaccine is a promising approach to cancer treatment. One of its strategies is to treat patient-derived tumor cells in vitro and then administer them in vivo to induce an adaptive immune response and achieve cancer treatment. Here, we want to explore the possibility of converting cancer tissue into a therapeutic tumor vaccine through induced immunogenic cell death (ICD) in situ. We loaded indocyanine green (ICG) into liposomes (ICG-Lipo) and modified it with the pardaxin peptide to realize an endoplasmic reticulum (ER)-targeting function (Par-ICG-Lipo). A microfluidic technique was developed for loading ICG, a water-soluble molecule, into liposomes with a high encapsulation efficiency (greater than 90%). Under near-infrared (NIR) irradiation, ER-targeting photodynamic therapy (PDT) induced by Par-ICG-Lipo could promote the release of danger-signaling molecules (DAMPs) and tumor antigens (TAAs) in vivo, which significantly enhanced the immunogenicity in vivo and thus stimulates a strong antitumor immune response. This process would be further amplified by adopting dendritic cells. In general, our strategy transformed in situ tumor cells into therapeutic vaccines by ER-targeting PDT, which could provide a clinically applicable and effective approach for cancer treatment.
Collapse
Affiliation(s)
- Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Xiang Li
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Jiaxin Huang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Bing Qin
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yongzhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 886 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| |
Collapse
|
3
|
Guo F, Hancock B, Griffith A, Lin H, Howard K, Keegan J, Zhang F, Chicoine A, Cahill L, Ng J, Lederer J. Distinct Injury Responsive Regulatory T Cells Identified by Multi-Dimensional Phenotyping. Front Immunol 2022; 13:833100. [PMID: 35634302 PMCID: PMC9135044 DOI: 10.3389/fimmu.2022.833100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/11/2022] [Indexed: 01/21/2023] Open
Abstract
CD4+ regulatory T cells (Tregs) activate and expand in response to different types of injuries, suggesting that they play a critical role in controlling the immune response to tissue and cell damage. This project used multi-dimensional profiling techniques to comprehensively characterize injury responsive Tregs in mice. We show that CD44high Tregs expand in response to injury and were highly suppressive when compared to CD44low Tregs. T cell receptor (TCR) repertoire analysis revealed that the CD44high Treg population undergo TCRαβ clonal expansion as well as increased TCR CDR3 diversity. Bulk RNA sequencing and single-cell RNA sequencing with paired TCR clonotype analysis identified unique differences between CD44high and CD44low Tregs and specific upregulation of genes in Tregs with expanded TCR clonotypes. Gene ontology analysis for molecular function of RNA sequencing data identified chemokine receptors and cell division as the most enriched functional terms in CD44high Tregs versus CD44low Tregs. Mass cytometry (CyTOF) analysis of Tregs from injured and uninjured mice verified protein expression of these genes on CD44high Tregs, with injury-induced increases in Helios, Galectin-3 and PYCARD expression. Taken together, these data indicate that injury triggers the expansion of a highly suppressive CD44high Treg population that is transcriptionally and phenotypically distinct from CD44low Tregs suggesting that they actively participate in controlling immune responses to injury and tissue damage.
Collapse
Affiliation(s)
- Fei Guo
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Brandon Hancock
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Alec Griffith
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Hui Lin
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Kaitlyn Howard
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Joshua Keegan
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Fan Zhang
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Adam Chicoine
- Human Immunology Center, Brigham and Women’s Hospital, Boston, MA, United States
| | - Laura Cahill
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Julie Ng
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - James Lederer
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Souza TFG, Pierdoná TM, Macedo FS, Aquino PEA, Rangel GFP, Duarte RS, Silva LMA, Viana GSB, Alves APNN, Montenegro RC, Wilke DV, Silveira ER, Alencar NMN. A proline derivative-enriched methanol fraction from Sideroxylon obtusifolium leaves (MFSOL) stimulates human keratinocyte cells and exerts a healing effect in a burn wound model. ACTA ACUST UNITED AC 2021; 54:e10700. [PMID: 34076141 PMCID: PMC8186379 DOI: 10.1590/1414-431x2021e10700] [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: 01/08/2021] [Accepted: 04/07/2021] [Indexed: 11/21/2022]
Abstract
It was previously demonstrated that the methanol fraction of Sideroxylon obtusifolium (MFSOL) promoted anti-inflammatory and healing activity in excisional wounds. Thus, the present work investigated the healing effects of MFSOL on human keratinocyte cells (HaCaT) and experimental burn model injuries. HaCaT cells were used to study MFSOL's effect on cell migration and proliferation rates. Female Swiss mice were subjected to a second-degree superficial burn protocol and divided into four treatment groups: Vehicle, 1.0% silver sulfadiazine, and 0.5 or 1.0% MFSOL Cream (CrMFSOL). Samples were collected to quantify the inflammatory mediators, and histological analyses were performed after 3, 7, and 14 days. The results showed that MFSOL (50 μg/mL) stimulated HaCaT cells by increasing proliferation and migration rates. Moreover, 0.5% CrMFSOL attenuated myeloperoxidase (MPO) activity and also stimulated the release of interleukin (IL)-1β and IL-10 after 3 days of treatment. CrMFSOL (0.5%) also enhanced wound contraction, promoted improvement of tissue remodeling, and increased collagen production after 7 days and VEGF release after 14 days. Therefore, MFSOL stimulated human keratinocyte (HaCaT) cells and improved wound healing via modulation of inflammatory mediators of burn injuries.
Collapse
Affiliation(s)
- T F G Souza
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T M Pierdoná
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil.,Faculty of Kinesiology and Recreation Management, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - F S Macedo
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P E A Aquino
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - G F P Rangel
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R S Duarte
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - L M A Silva
- Embrapa Agroindustria Tropical, Fortaleza, CE, Brasil
| | - G S B Viana
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A P N N Alves
- Departamento de Clínica Odontológica, Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R C Montenegro
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - D V Wilke
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - E R Silveira
- Departamento de Química Orgânica e Inorgânica, Centro de Ciências, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - N M N Alencar
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
5
|
Yamakawa K, Tajima G, Keegan JW, Nakahori Y, Guo F, Seshadri AJ, Cahill LA, Lederer JA. Trauma induces expansion and activation of a memory-like Treg population. J Leukoc Biol 2021; 109:645-656. [PMID: 32531832 PMCID: PMC10228755 DOI: 10.1002/jlb.4a0520-122r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/30/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022] Open
Abstract
CD4+ regulatory T cells (Tregs) are acutely activated by traumatic injury, which suggests that they may react to injury with similar kinetics as memory T cells. Here, we used a mouse burn trauma model to screen for memory-like T cell responses to injury by transferring T cells from sham or burn CD45.1 mice into CD45.2 mice and performing secondary injuries in recipient mice. Among all T cell subsets that were measured, only Tregs expanded in response to secondary injury. The expanded Tregs were a CD44high /CD62Llow subpopulation, markers indicative of memory T cells. CyTOF (cytometry by time-of-flight) mass cytometry was used to demonstrate that injury-expanded Tregs expressed higher levels of CD44, CTLA-4, ICOS, GITR, and Helios than Tregs from noninjured mice. Next, we tested whether a similar population of Tregs might react acutely to burn trauma. We observed that Tregs with a phenotype that matched the injury-expanded Tregs were activated by 6 h after injury. To test if Treg activation by trauma requires functional MHC class II, we measured trauma-induced Treg activation in MHC class II gene deficient (MHCII-/- ) mice or in mice that were given Fab fragment of anti-MHC class II antibody to block TCR activation. Injury-induced Treg activation occurred in normal mice but only partial activation was detected in MHCII-/- mice or in mice that were given Fab anti-MHCII antibody. These findings demonstrate that trauma activates a memory-like Treg subpopulation and that Treg activation by injury is partially dependent on TCR signaling by an MHC class II dependent mechanism.
Collapse
Affiliation(s)
- Kazuma Yamakawa
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Goro Tajima
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Emergency Medicine, Unit of Clinical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Joshua W. Keegan
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yasutaka Nakahori
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Fei Guo
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anupamaa J. Seshadri
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Laura A. Cahill
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - James A. Lederer
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Yadav PK, Chandrakar P, Sharma P, Vishwakarma P, Parmar N, Srivastava M, Kar S. Reciprocal changes in CD11c +CD11b + and CD11c +CD8α + dendritic cell subsets determine protective or permissive immune response in murine experimental VL. Vaccine 2020; 38:355-365. [PMID: 31648908 DOI: 10.1016/j.vaccine.2019.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 01/02/2023]
Abstract
CD11c+CD8α+ and CD11c+CD11b+ dendritic cells are two major subsets of murine splenic CD11c+ DCs which play a crucial role in T cell priming and shaping Th1/Th2 responses, but their role in the context of experimental visceral leishmaniasis (VL) is poorly understood. Herein, we showed that L. donovani infection in Balb/c mice preferentially decreased the population abundance of CD11c+CD11b+ DCs and increased relative abundance of splenic CD11c+CD8α +DCs. During infection, splenic CD11c+CD11b+ DCs induced Th1 differentiation whereas CD11c+CD8α+ DCs promoted Th2 differentiation. Additionally, treatment of infected mice with miltefosine as experimental control exhibited host defense allowing the restoration of CD11c+CD11b+ population and decrease in CD11c+CD8α+ subset. Furthermore, reciprocal regulation of immune accessory surface molecules, Sema4A and OX40L critically determined Th1/Th2 response induced by these DC subsets during VL. L. donovani infection significantly induced OX40L expression and slightly downregulated SEMA 4A expression in CD11c+CD8α+ DCs whereas miltefosine treatment significantly downregulated OX40L expression along with pronounced upregulation of SEMA 4A expression in CD11c+CD11b+ DCs. SiRNA mediated knockdown of SEMA 4A markedly reduced CD11c+CD11b+ driven IFN-γ, TNF-α and IL-12 synthesis in miltefosine treated mice whereas functional blocking of OX40L decreased CD11c+CD8α+ induced IL-10, IL-4 and TGF-β synthesis in L. donovani infected group. Vaccination of Balb/c mice with antigen-pulsed + CpG-ODN-activated DC subsets revealed that only antigen-pulsed CD11c+CD11b+ DCs eliminated parasite load in visceral organ and restored protective Th1 cytokine response. Collectively, our results suggest that differential regulation of splenic CD11c+ subsets by L. donovani is essential for disease progression and specific subtypes may be exploited as prophylactic measures against visceral leishmaniasis.
Collapse
Affiliation(s)
- Pawan Kumar Yadav
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, New Delhi, India
| | - Pragya Chandrakar
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, New Delhi, India
| | - Pankaj Sharma
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Preeti Vishwakarma
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, New Delhi, India
| | - Naveen Parmar
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, New Delhi, India
| | - Mrigank Srivastava
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, New Delhi, India
| | - Susanta Kar
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, New Delhi, India.
| |
Collapse
|
7
|
Zhang X, Li N, Meng Y, Zhang R, Bian J, Yao Y, Li J, Deng X. High-Level Expression of Toll-Like Receptors on Dendritic Cells in Adult Patients with Burns on ≥90% of Total Body Surface Area (TBSA). Med Sci Monit 2016; 22:3493-3499. [PMID: 27686145 PMCID: PMC5047034 DOI: 10.12659/msm.897433] [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] [Indexed: 11/30/2022] Open
Abstract
Background As a serious clinical problem, severe burn injury disturbs the immune system, resulting in progressive suppression of immune response. TLRs are associated with immune system activation, but the effect of TLRs levels on circulating cDCs of severe burn injury patients has not been fully assessed. Material/Methods Ten patients with total body surface area (TBSA) burned >90% admitted to our institution were enrolled in this study. We analyzed TLR2, TLR4, and TLR9 expression on the circulating cDCs by using multicolor flow cytometric analysis in patients at 14 days to 28 days after burn injury according to mortality, and We also assessed Demographics, clinical outcomes, organ function, and inflammatory and acute-phase responses. Results No difference in TBSA, sex, age, or number of operations before the first 14 days after injury were observed between surviving and non-surviving burn patients. The levels of TLR2, TLR4, and TLR9 in circulating cDCs were significantly and consistently elevated in all patients compared to age-matched healthy volunteers, and survivors exhibited higher TLR2 and TLR4 values than non-survivors. Of the survivors, TLR2 and TLR4 levels were higher at 28 days than at 14 days after injury, while the difference in TLR9 levels was not significant. TLR2 levels of non-survivors at 28 days after injury decreased, and the TLR4 and TLR9 levels showed no significant difference. Conclusions TLRs levels in circulating cDCs are highly activated in severe burn injury patients up to 28 days after injury. The low expression of TLR2 in cDCs may be useful as a potential marker predicting the poor prognosis of severe burn patients.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Na Li
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Yan Meng
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Renjing Zhang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Jinjun Bian
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Ying Yao
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Jinbao Li
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| | - Xiaoming Deng
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China (mainland)
| |
Collapse
|
8
|
Abstract
Traumatic injury remains one of the most prevalent reasons for patients to be hospitalized. Burn injury accounts for 40,000 hospitalizations in the United States annually, resulting in a large burden on both the health and economic system and costing millions of dollars every year. The complications associated with postburn care can quickly cause life-threatening conditions including sepsis and multiple organ dysfunction and failure. In addition, alcohol intoxication at the time of burn injury has been shown to exacerbate these problems. One of the biggest reasons for the onset of these complications is the global suppression of the host immune system and increased susceptibility to infection. It has been hypothesized that infections after burn and other traumatic injury may stem from pathogenic bacteria from within the host's gastrointestinal tract. The intestine is the major reservoir of bacteria within the host, and many studies have demonstrated perturbations of the intestinal barrier after burn injury. This article reviews the findings of these studies as they pertain to changes in the intestinal immune system after alcohol and burn injury.
Collapse
|
9
|
Noronha SAACD, Noronha SMRD, Lanziani LE, Ferreira LM, Gragnani A. Innate and adaptive immunity gene expression of human keratinocytes cultured of severe burn injury. Acta Cir Bras 2015; 29 Suppl 3:60-7. [PMID: 25351159 DOI: 10.1590/s0102-86502014001700012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Evaluate the expression profile of genes related to Innate and Adaptive Immune System (IAIS) of human Primary Epidermal keratinocytes (hPEKP) of patients with severe burns. METHODS After obtaining viable fragments of skin with and without burning, culture hKEP was initiated by the enzymatic method using Dispase (Sigma-Aldrich). These cells were treated with Trizol(r) (Life Technologies) for extraction of total RNA. This was quantified and analyzed for purity for obtaining cDNA for the analysis of gene expression using specific IAIS PCR Arrays plates (SA Biosciences). RESULTS After the analysis of gene expression we found that 63% of these genes were differentially expressed, of which 77% were repressed and 23% were hyper-regulated. Among these, the following genes (fold increase or decrease): IL8 (41), IL6 (32), TNF (-92), HLA-E (-86), LYS (-74), CCR6 (- 73), CD86 (-41) and HLA-A (-35). CONCLUSIONS This study contributes to the understanding of the molecular mechanisms underlying wound infection caused by the burn. Furthermore, it may provide new strategies to restore normal expression of these genes and thereby change the healing process and improve clinical outcome.
Collapse
Affiliation(s)
| | | | | | | | - Alfredo Gragnani
- Department of Surgery, Paulista School of Medicine, Federal University of Sao Paulo, Brazil
| |
Collapse
|
10
|
Fazal N. OX62+OX6+OX35+ rat dendritic cells are unable to prime CD4+ T cells for an effective immune response following acute burn injury. RESULTS IN IMMUNOLOGY 2013; 3:64-72. [PMID: 24600560 DOI: 10.1016/j.rinim.2013.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/22/2013] [Accepted: 06/24/2013] [Indexed: 02/02/2023]
Abstract
Co-stimulatory molecules expressed on Dendritic Cells (DCs) function to coordinate an efficient immune response by T cells in the peripheral lymph nodes. We hypothesized that CD4+ T cell-mediated immune suppression following burn injury may be related to dysfunctional DCs residing in gut associated lymphoid tissues (GALT), such as Mesenteric Lymph Nodes (MLN). Therefore, we studied co-stimulatory molecules expressed on burn rat MLN DCs as an index of functional DCs that would mount an effective normal CD4+ T cell immune response. In a rat model of 30% Total Body Surface Area (TBSA) scald burn, OX62+OX6+OX35+ DCs and CD4+ T cells were isolated from MLN of day 3 post-burn and sham control rats. DCs were tested for their expression of co-stimulatory molecules, and prime CD4+ T cell (DC:CD4+T cell co-culture assays) to determine an effector immune response such as CD4+ T cell proliferation. The surface receptor expressions of MLN DCs co-stimulatory molecules, i.e., MHC-II, CD40, CD80 (B7-1), and CD86 (B7-2) were determined by Flow cytometry (quantitatively) and confocal microscopy (qualitatively). Tritiated thymidine and CFDA-SE determined CD4+ T cell proliferation following co-incubation with DCs. Cytokine milieu of MLN (IL-12 and IL-10) was assessed by mRNA determination by RT-PCR. The results showed down-regulated expressions of co-stimulatory markers (CD80, CD86, CD40 and MHC-II) of MLN DCs obtained from burn-injured rats, as well as lack of ability of these burn-induced DCs to stimulate CD4+ T cell proliferation in co-culture assays, as compared to the sham rats. Moreover, anti-CD40 stimulation of affected burn MLN DCs did not reverse this alteration. Furthermore, a marked up-regulation of mRNA IL-10 and down-regulation of mRNA IL-12 in burn MLN as compared to sham animals was also observed. To surmise, the data indicated that dysfunctional OX62+OX6+OX35+ rat MLN DCs may contribute to CD4+ T-cell-mediated immune suppression observed following acute burn injury.
Collapse
Affiliation(s)
- Nadeem Fazal
- Department of Pharmaceutical Sciences, College of Pharmacy, 206 Douglas Hall, Chicago State University, 9501 South King Drive, Chicago, IL 60628, USA
| |
Collapse
|
11
|
Burn injury triggered dysfunction in dendritic cell response to TLR9 activation and resulted in skewed T cell functions. PLoS One 2012. [PMID: 23189191 PMCID: PMC3506591 DOI: 10.1371/journal.pone.0050238] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Severe trauma such as burn injury is often associated with a systemic inflammatory syndrome characterized by a hyperactive innate immune response and suppressed adaptive immune function. Dendritic cells (DCs), which sense pathogens via their Toll-like receptors (TLRs), play a pivotal role in protecting the host against infections. The effect of burn injury on TLR-mediated DC function is a debated topic and the mechanism controlling the purported immunosuppressive response remains to be elucidated. Here we examined the effects of burn injury on splenic conventional DC (cDC) and plasmacytoid DC (pDC) responses to TLR9 activation. We demonstrate that, following burn trauma, splenic cDCs' cytokine production profile in response to TLR9 activation became anti-inflammatory dominant, with high production of IL-10 (>50% increase) and low production of IL-6, TNF-α and IL-12p70 (∼25-60% reduction). CD4+ T cells activated by these cDCs were defective in producing Th1 and Th17 cytokines. Furthermore, burn injury had a more accentuated effect on pDCs than on cDCs. Following TLR9 activation, pDCs displayed an immature phenotype with an impaired ability to secrete pro-inflammatory cytokines (IFN-α, IL-6 and TNF-α) and to activate T cell proliferation. Moreover, cDCs and pDCs from burn-injured mice had low transcript levels of TLR9 and several key molecules of the TLR signaling pathway. Although hyperactive innate immune response has been associated with severe injury, our data show to the contrary that DCs, as a key player in the innate immune system, had impaired TLR9 reactivity, an anti-inflammatory phenotype, and a dysfunctional T cell-priming ability. We conclude that burn injury induced impairments in DC immunobiology resulting in suppression of adaptive immune response. Targeted DC immunotherapies to promote their ability in triggering T cell immunity may represent a strategy to improve immune defenses against infection following burn injury.
Collapse
|
12
|
Abstract
The inflammasome is activated in response to pathogen or endogenous danger signals and acts as an initiator and mediator of inflammatory reactions. In this study, we wished to identify whether the inflammasome is activated in vivo by injury. And if so, we wanted to characterize the kinetics, the immune cell distribution, and the functional impact of inflammasome activation on the injury response. Because caspase-1 activation is the final product of the inflammasome pathway, we used cleaved caspase-1 p10 and p20 as a measure for inflammasome activation in cells. We first developed a procedure to stain for caspase-1 p10 and p20 by flow cytometry (FACS) in lipopolysaccharide + adenosine triphosphate-stimulated spleen cells. This method for measuring caspase-1 activation was validated using FLICA (fluorochrome inhibitor of caspase), a fluorescently tagged specific binding reagent for activated caspase-1. Once validated by in vitro studies, we measured caspase-1 activation by FACS in immune cell subsets prepared from the lymph nodes and spleens of sham- or burn-injured mice at different time points. Lastly, the functional significance of inflammasome activation following burn injury was tested in mice treated with the specific caspase-1 inhibitor, AC-YVAD-CMK. The results of in vitro studies indicated that adenosine triphosphate and lipopolysaccharide stimulation induced significant caspase-1 activation in dendritic cells, macrophages, and natural killer (NK) cells. This approach also revealed caspase-1 activation in CD4 and CD8 T cells as well as B cells. We then measured caspase-1 activation in cells prepared from the lymph nodes and spleens of sham- or burn-injured mice. Significant caspase-1 activation was detected in macrophages and dendritic cells by 4 h after injury and peaked by day 1 after injury. FLICA staining confirmed that caspase-1 activation occurred in these cells at 1 day after injury. We also found significant injury-induced caspase-1 activation in NK cells, CD4 T cells, and B cells, but CD8 T cells did not demonstrate caspase-1 activation. Surprisingly, we found that blocking caspase-1 activation with AC-YVAD-CMK in vivo caused significantly higher mortality in burn-injured mice (P < 0.01). Taken together, these findings document that injury induces inflammasome activation in many immune cell subsets, but primarily in macrophages, and that inflammasome activation plays a protective role in the host response to severe injury.
Collapse
|
13
|
Howell K, Posluszny J, He LK, Szilagyi A, Halerz J, Gamelli RL, Shankar R, Muthu K. High MafB expression following burn augments monocyte commitment and inhibits DC differentiation in hemopoietic progenitors. J Leukoc Biol 2011; 91:69-81. [PMID: 21984745 DOI: 10.1189/jlb.0711338] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We have previously shown that perturbed bone marrow progenitor development promotes hyporesponsive monocytes following experimental burn sepsis. Clinical and experimental sepsis is associated with monocyte deactivation and depletion of mDCs. Decrease in circulating DCs is reported in burn patients who develop sepsis. In our 15% TBSA scald burn model, we demonstrate a significant reduction in the circulating MHC-II(+) population and mDCs (Gr1(neg)CD11b(+)CD11c(+)) with a corresponding decrease in bone marrow MHC-II(+) cells and mDCs for up to 14 days following burn. We explored the underlying mechanism(s) that regulate bone marrow development of monocytes and DCs following burn injury. We found a robust bone marrow response with a significant increase in multipotential HSCs (LSK) and bipotential GMPs following burn injury. GMPs from burn mice exhibit a significant reduction in GATA-1, which is essential for DC development, but express high levels of MafB and M-CSFRs, both associated with monocyte production. GMPs obtained from burn mice differentiated 1.7 times more into Mϕ and 1.6-fold less into DCs compared with sham. Monocytes and DCs expressed 50% less MHC-II in burn versus sham. Increased monocyte commitment in burn GMPs was a result of high MafB and M-CSFR expressions. Transient silencing of MafB (siRNA) in GMP-derived monocytes from burn mice partially restored DC differentiation deficits and increased GATA-1 expression. We provide evidence that high MafB following burn plays an inhibitory role in monocyte-derived DC differentiation by regulating M-CSFR and GATA-1 expressions.
Collapse
Affiliation(s)
- Kirstin Howell
- Department of Surgery, Mount Sinai Medical Center, Chicago, Illinois, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Hanschen M, Tajima G, O’Leary F, Ikeda K, Lederer JA. Injury induces early activation of T-cell receptor signaling pathways in CD4+ regulatory T cells. Shock 2011; 35:252-7. [PMID: 20720513 PMCID: PMC3045756 DOI: 10.1097/shk.0b013e3181f489c5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although it is known that injury enhances the regulatory activity of CD4 regulatory T cells (Tregs), the cellular and molecular mechanisms responsible for injury-induced Treg activation remain unclear. This study was designed to investigate and compare injury-induced T-cell receptor (TCR) signaling in Tregs, non-Tregs, and CD8 T cells. Specifically, we used phospho-flow cytometry to measure the expression and phosphorylation of ZAP-70, protein kinase C θ, nuclear factor of activated T cells, and glycogen synthase kinase 3β in FoxP3 Tregs versus FoxP3 non-Tregs versus CD8 T cells. Groups of male C57BL/6J mice underwent burn or sham injury, and lymph nodes and spleens were harvested at early time points-15, 30, 60, 120, and 240 min-to measure TCR signaling. As early as 15 min after burn injury, we observed a significant upregulation and phosphorylation of ZAP-70, protein kinase C θ, nuclear factor of activated T cells, and glycogen synthase kinase 3β in Tregs prepared from injury-site-draining lymph nodes. Burn injury did not activate TCR signaling in Tregs from the spleen or in CD4 non-Tregs and CD8 T cells. In conclusion, the results of this study demonstrate that burn injury activates TCR signaling in Tregs, but not non-Tregs or CD8 T cells. These findings suggest that injury provides an early TCR-activating signal to Tregs and supply new insights into how injury influences the adaptive immune system.
Collapse
Affiliation(s)
- Marc Hanschen
- Department of Surgery (Immunology), Brigham and Women’s Hospital/Harvard Medical School, Boston, MA
- Department of Trauma and Orthopaedic Surgery, Ludwig-Maximilians-University, Munich, Germany
| | - Goro Tajima
- Department of Surgery (Immunology), Brigham and Women’s Hospital/Harvard Medical School, Boston, MA
| | - Fionnuala O’Leary
- Department of Surgery (Immunology), Brigham and Women’s Hospital/Harvard Medical School, Boston, MA
| | - Kimiko Ikeda
- Department of Surgery (Immunology), Brigham and Women’s Hospital/Harvard Medical School, Boston, MA
| | - James A. Lederer
- Department of Surgery (Immunology), Brigham and Women’s Hospital/Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
van den Berg LM, de Jong MAWP, Witte LD, Ulrich MMW, Geijtenbeek TBH. Burn injury suppresses human dermal dendritic cell and Langerhans cell function. Cell Immunol 2011; 268:29-36. [PMID: 21345420 DOI: 10.1016/j.cellimm.2011.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 01/28/2011] [Accepted: 01/28/2011] [Indexed: 10/18/2022]
Abstract
Human skin contains epidermal Langerhans cells (LCs) and dermal dendritic cells (DCs) that are key players in induction of adaptive immunity upon infection. After major burn injury, suppressed adaptive immunity has been observed in patients. Here we demonstrate that burn injury affects adaptive immunity by altering both epidermal LC and dermal DC functions. We developed a human ex vivo burn injury model to study the function of DCs in thermally injured skin. No differences were observed in the capacity of both LCs and dermal DCs to migrate out of burned skin compared to unburned skin. Similarly, expression levels of co-stimulatory molecules were unaltered. Notably, we observed a strong reduction of T cell activation induced by antigen presenting cell (APC) subsets that migrated from burned skin through soluble burn factors. Further analyses demonstrated that both epidermal LCs and dermal DCs have a decreased T cell stimulatory capacity after burn injury. Restoring the T cell stimulatory capacity of DC subsets might improve tissue regeneration in patients with burn wounds.
Collapse
Affiliation(s)
- Linda M van den Berg
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
16
|
Suzuki T, Kawasaki T, Choudhry MA, Chaudry IH. Role of PPARγ in the salutary effects of 17β-estradiol on Kupffer cell cytokine production following trauma-hemorrhage. J Cell Physiol 2010; 226:205-11. [PMID: 20665707 DOI: 10.1002/jcp.22327] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Studies have shown that administration of 17β-estradiol prevents trauma-hemorrhage-induced increase in proinflammatory cytokine production by Kupffer cells and associated multiple organ injury. Since activation of peroxisome proliferator-activated receptor γ (PPARγ) following ischemic conditions has been shown to be protective, we examined if PPARγ plays any role in the salutary effects of 17β-estradiol on Kupffer cell cytokine production following trauma-hemorrhage. Male mice underwent trauma-hemorrhage (mean blood pressure 40 mmHg for 90 min, then resuscitation). 17β-estradiol (50 µg/kg) or vehicle with or without PPARγ antagonist GW9662 was injected subcutaneously at the middle of resuscitation. At 2 h after trauma-hemorrhage, plasma interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels, Kupffer cell IL-6 and TNF-α production and mRNA expression, and PPARγ, nuclear factor (NF)-κB and activator protein (AP)-1 DNA binding activity were determined. Kupffer cell IL-6 and TNF-α production, as well as plasma IL-6 and TNF-α levels, increased following trauma-hemorrhage. Moreover, NF-κB and AP-1 DNA binding activity and IL-6 and TNF-α mRNA expression were also enhanced under such conditions. However, 17β-estradiol administration normalized all these parameters. Although PPARγ activity decreased after trauma-hemorrhage, administration of 17β-estradiol following trauma-hemorrhage elevated PPARγ activity above the normal level. Inhibition of PPARγ by co-administration of GW9662, however, abolished the salutary effects of 17β-estradiol on plasma cytokine and Kupffer cells. Thus, activation of PPARγ appears to play an important role in mediating the salutary effects of 17β-estradiol on plasma cytokine levels and Kupffer cell cytokine production after trauma-hemorrhage, which are likely mediated via NF-κB and AP-1.
Collapse
Affiliation(s)
- Takao Suzuki
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | | | |
Collapse
|