1
|
Tian Y, Huang S, Zhou G, Fei X, Li YA, Li Q, Wang S, Shi H. Evaluation of immunogenicity and protective efficacy of outer membrane vesicles from Salmonella Typhimurium and Salmonella Choleraesuis. Vet Microbiol 2024; 294:110131. [PMID: 38805917 DOI: 10.1016/j.vetmic.2024.110131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
Outer membrane vesicles (OMVs) are membranous structures frequently observed in Gram-negative bacteria that contain bioactive substances. These vesicles are rich in bacterial antigens that can activate the host's immune system, making them a promising candidate vaccine to prevent and manage bacterial infections. The aim of this study was to assess the immunogenicity and protective efficacy of OMVs derived from Salmonella enterica serovar Typhimurium and S. Choleraesuis, while also focusing on enhancing OMV production. Initial experiments showed that OMVs from wild-type strains did not provide complete protection against homologous Salmonella challenge, possible due to the presence of flagella in the purified OMVs samples, which may elicit an unnecessary immune response. To address this, flagellin-deficient mutants of S. Typhimurium and S. Choleraesuis were constructed, designated rSC0196 and rSC0199, respectively. These mutants exhibited reduced cell motility and their OMVs were found to be flagellin-free. Immunization with non-flagellin OMVs derived from rSC0196 induced robust antibody responses and improved survival rates in mice, as compared to the OMVs derived from the wild-type UK-1. In order to enhance OMV production, deletions of ompA or tolR were introduced into rSC0196. The deletion of tolR not only increase the yield of OMVs, but also conferred complete protection against homologous S. Typhimurium challenge in mice. Collectively, these findings indicate that the flagellin-deficient OMVs with a tolR mutation have the potential to serve as a versatile vaccine platform, capable of inducing broad-spectrum protection against significant pathogens.
Collapse
Affiliation(s)
- Yichen Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Shan Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Xia Fei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Yu-An Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China.
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611-0880, USA
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety (JIRLAAPS), Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
2
|
Mulder PPG, Hooijmans CR, Vlig M, Middelkoop E, Joosten I, Koenen HJPM, Boekema BKHL. Kinetics of Inflammatory Mediators in the Immune Response to Burn Injury: Systematic Review and Meta-Analysis of Animal Studies. J Invest Dermatol 2024; 144:669-696.e10. [PMID: 37806443 DOI: 10.1016/j.jid.2023.09.269] [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: 05/25/2023] [Revised: 08/31/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
Burns are often accompanied by a dysfunctional immune response, which can lead to systemic inflammation, shock, and excessive scarring. The objective of this study was to provide insight into inflammatory pathways associated with burn-related complications. Because detailed information on the various inflammatory mediators is scattered over individual studies, we systematically reviewed animal experimental data for all reported inflammatory mediators. Meta-analyses of 352 studies revealed a strong increase in cytokines, chemokines, and growth factors, particularly 19 mediators in blood and 12 in burn tissue. Temporal kinetics showed long-lasting surges of proinflammatory cytokines in blood and burn tissue. Significant time-dependent effects were seen for IL-1β, IL-6, TGF-β1, and CCL2. The response of anti-inflammatory mediators was limited. Burn technique had a profound impact on systemic response levels. Large burn size and scalds further increased systemic, but not local inflammation. Animal characteristics greatly affected inflammation, for example, IL-1β, IL-6, and TNF-α levels were highest in young, male rats. Time-dependent effects and dissimilarities in response demonstrate the importance of appropriate study design. Collectively, this review presents a general overview of the burn-induced immune response exposing inflammatory pathways that could be targeted through immunotherapy for burn patients and provides guidance for experimental set-ups to advance burn research.
Collapse
Affiliation(s)
- Patrick P G Mulder
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Carlijn R Hooijmans
- Meta-Research Team, Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel Vlig
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands
| | - Esther Middelkoop
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Tissue Function and Regeneration, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bouke K H L Boekema
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Niu Z, Wei G, Liang H, Wang X, Yang W, Wei G, Guo J, Chen Y, Tao R, Niu J. Bioinformatics-Led Identification of Potential Biomarkers and Inflammatory Infiltrates in Burn Injury. J Burn Care Res 2023; 44:1382-1392. [PMID: 37022972 DOI: 10.1093/jbcr/irad050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Indexed: 04/07/2023]
Abstract
Burn injury is a life-threatening disease with a poor prognosis. The immune change and underlying mechanisms remain largely unknown. Thus, this study aims to find potential biomarkers and analyze the immune infiltrates after burn injury. Gene expression data of burn patients were obtained from the Gene Expression Omnibus database. Key immune-related genes (IRGs) were screened by differential and least absolute shrinkage and selection operator (LASSO) regression analysis. Based on key IRGs, patients were divided into two clusters by consensus cluster analysis. Immune infiltration was analyzed by the single sample gene set enrichment analysis (GSEA) method and the immune score was calculated by the principal component analysis method. A nomogram model was constructed based on the calculated immune score and clinical features. Finally, the expression of screened key genes was validated by an external cohort and quantitative polymerase chain reaction experiment. Fifty-nine IRGs were differently expressed in burn patients. After LASSO regression analysis, 12 key genes remained, namely AZU1, OLR1, RNASE2, FGF13, NR1D2, NR2E1, TLR5, CAMP, DEFA4, PGLYRP1, CTSG, and CCR3. Then, patients were divided into two clusters. Immune infiltration analysis revealed that more immune cells were infiltrated and more pathways were activated in cluster A, in which patients showed high immune scores. Finally, a nomogram model was constructed and showed high accuracy and reliability. The expression pattern of 12 key genes in an external cohort and clinical samples was in accordance with the theoretical analysis results. In conclusion, this research elucidated the key role of immune response in burns and could be used as a guide for burn treatment.
Collapse
Affiliation(s)
- Zehao Niu
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guoxing Wei
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Hao Liang
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xin Wang
- PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Wenjuan Yang
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Gang Wei
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiachang Guo
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingen Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jun Niu
- Department of Burns and Plastic surgery, The 83 Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
4
|
Schwabkey ZI, Wiesnoski DH, Chang CC, Tsai WB, Pham D, Ahmed SS, Hayase T, Turrubiates MRO, El-Himri RK, Sanchez CA, Hayase E, Oquendo ACF, Miyama T, Halsey TM, Heckel BE, Brown AN, Jin Y, Raybaud M, Prasad R, Flores I, McDaniel L, Chapa V, Lorenzi PL, Warmoes MO, Tan L, Swennes AG, Fowler S, Conner M, McHugh K, Graf T, Jensen VB, Peterson CB, Do KA, Zhang L, Shi Y, Wang Y, Galloway-Pena JR, Okhuysen PC, Daniel-MacDougall CR, Shono Y, da Silva MB, Peled JU, van den Brink MR, Ajami N, Wargo JA, Reddy P, Valdivia RH, Davey L, Rondon G, Srour SA, Mehta RS, Alousi AM, Shpall EJ, Champlin RE, Shelburne SA, Molldrem JJ, Jamal MA, Karmouch JL, Jenq RR. Diet-derived metabolites and mucus link the gut microbiome to fever after cytotoxic cancer treatment. Sci Transl Med 2022; 14:eabo3445. [PMID: 36383683 PMCID: PMC10028729 DOI: 10.1126/scitranslmed.abo3445] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Not all patients with cancer and severe neutropenia develop fever, and the fecal microbiome may play a role. In a single-center study of patients undergoing hematopoietic cell transplant (n = 119), the fecal microbiome was characterized at onset of severe neutropenia. A total of 63 patients (53%) developed a subsequent fever, and their fecal microbiome displayed increased relative abundances of Akkermansia muciniphila, a species of mucin-degrading bacteria (P = 0.006, corrected for multiple comparisons). Two therapies that induce neutropenia, irradiation and melphalan, similarly expanded A. muciniphila and additionally thinned the colonic mucus layer in mice. Caloric restriction of unirradiated mice also expanded A. muciniphila and thinned the colonic mucus layer. Antibiotic treatment to eradicate A. muciniphila before caloric restriction preserved colonic mucus, whereas A. muciniphila reintroduction restored mucus thinning. Caloric restriction of unirradiated mice raised colonic luminal pH and reduced acetate, propionate, and butyrate. Culturing A. muciniphila in vitro with propionate reduced utilization of mucin as well as of fucose. Treating irradiated mice with an antibiotic targeting A. muciniphila or propionate preserved the mucus layer, suppressed translocation of flagellin, reduced inflammatory cytokines in the colon, and improved thermoregulation. These results suggest that diet, metabolites, and colonic mucus link the microbiome to neutropenic fever and may guide future microbiome-based preventive strategies.
Collapse
Affiliation(s)
- Zaker I. Schwabkey
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Diana H. Wiesnoski
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chia-Chi Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wen-Bin Tsai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dung Pham
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Saira S. Ahmed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tomo Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Rawan K. El-Himri
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher A. Sanchez
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eiko Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Annette C. Frenk Oquendo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Takahiko Miyama
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Taylor M. Halsey
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Brooke E. Heckel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexandria N. Brown
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yimei Jin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mathilde Raybaud
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rishika Prasad
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ivonne Flores
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lauren McDaniel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Valerie Chapa
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marc O. Warmoes
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alton G. Swennes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephanie Fowler
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Margaret Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin McHugh
- CPRIT Scholar in Cancer Research, Austin, TX 78701, USA
- Department of Bioengineering, Rice University, Houston, TX 77251, USA
| | - Tyler Graf
- Department of Bioengineering, Rice University, Houston, TX 77251, USA
| | - Vanessa B. Jensen
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine B. Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Liangliang Zhang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yushu Shi
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jessica R. Galloway-Pena
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA
| | - Pablo C. Okhuysen
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Yusuke Shono
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marina Burgos da Silva
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan U. Peled
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10021, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marcel R.M. van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10021, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nadim Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A. Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pavan Reddy
- Department of Hematology and Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Raphael H. Valdivia
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710 USA
| | - Lauren Davey
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710 USA
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samer A. Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rohtesh S. Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amin M. Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard E. Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samuel A. Shelburne
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey J. Molldrem
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mohamed A. Jamal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L. Karmouch
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert R. Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- CPRIT Scholar in Cancer Research, Austin, TX 78701, USA
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Wang P, Zhang Z, Yin B, Li J, Xialin C, Lian W, Su Y, Jia C. Identifying changes in immune cells and constructing prognostic models using immune-related genes in post-burn immunosuppression. PeerJ 2022; 10:e12680. [PMID: 35070500 PMCID: PMC8761370 DOI: 10.7717/peerj.12680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/02/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Burn patients are prone to infection as well as immunosuppression, which is a significant cause of death. Currently, there is a lack of prognostic biomarkers for immunosuppression in burn patients. This study was conducted to identify immune-related genes that are prognosis biomarkers in post-burn immunosuppression and potential targets for immunotherapy. METHODS We downloaded the gene expression profiles and clinical data of 213 burn patients and 79 healthy samples from the Gene Expression Omnibus (GEO) database. Immune infiltration analysis was used to identify the proportion of circulating immune cells. Functional enrichment analyses were carried out to identify immune-related genes that were used to build miRNA-mRNA networks to screen key genes. Next, we carried out correlation analysis between immune cells and key genes that were then used to construct logistic regression models in GSE77791 and were validated in GSE19743. Finally, we determined the expression of key genes in burn patients using quantitative reverse transcription polymerase chain reaction (qRT-PCR). RESULTS A total of 745 differently expressed genes were screened out: 299 were up-regulated and 446 were down-regulated. The number of Th-cells (CD4+) decreased while neutrophils increased in burn patients. The enrichment analysis showed that down-regulated genes were enriched in the T-cell activation pathway, while up-regulated genes were enriched in neutrophil activation response in burn patients. We screened out key genes (NFATC2, RORA, and CAMK4) that could be regulated by miRNA. The expression of key genes was related to the proportion of Th-cells (CD4+) and survival, and was an excellent predictor of prognosis in burns with an area under the curve (AUC) value of 0.945. Finally, we determined that NFATC2, RORA, and CAMK4 were down-regulated in burn patients. CONCLUSION We found that NFATC2, RORA, and CAMK4 were likely prognostic biomarkers in post-burn immunosuppression and potential immunotherapeutic targets to convert Th-cell dysfunction.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zexin Zhang
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bin Yin
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiayuan Li
- Department of Anesthesia Operation, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Cheng Xialin
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenqin Lian
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yingjun Su
- Department of Burns and Plastic Surgery, Plastic Surgery Hospital, Xi’an International Medical Center, Xi’an, Shaanxi, China
| | - Chiyu Jia
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
6
|
de Maria YNLF, Aciole Barbosa D, Menegidio FB, Santos KBNH, Humberto AC, Alencar VC, Silva JFS, Costa de Oliveira R, Batista ML, Nunes LR, Jabes DL. Analysis of mouse faecal dysbiosis, during the development of cachexia, induced by transplantation with Lewis lung carcinoma cells. MICROBIOLOGY (READING, ENGLAND) 2021; 167. [PMID: 34596506 DOI: 10.1099/mic.0.001088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cachexia (CC) is a complex wasting syndrome that significantly affects life quality and life expectancy among cancer patients. Original studies, in which CC was induced in mouse models through inoculation with BaF and C26 tumour cells, demonstrated that CC development correlates with bacterial gut dysbiosis in these animals. In both cases, a common microbial signature was observed, based on the expansion of Enterobacteriaceae in the gut of CC animals. However, these two types of tumours induce unique microbial profiles, suggesting that different CC induction mechanisms significantly impact the outcome of gut dysbiosis. The present study sought to expand the scope of such analyses by characterizing the CC-associated dysbiosis that develops when mice are inoculated with Lewis lung carcinoma (LLC) cells, which constitutes one of the most widely employed mechanisms for CC induction. Interestingly, Enterobacteriaceae expansion is also observed in LLC-induced CC. However, the dysbiosis identified herein displays a more complex pattern, involving representatives from seven different bacterial phyla, which were consistently identified across successive levels of taxonomic hierarchy. These results are supported by a predictive analysis of gene content, which identified a series of functional/structural changes that potentially occur in the gut bacterial population of these animals, providing a complementary and alternative approach to microbiome analyses based solely on taxonomic classification.
Collapse
Affiliation(s)
- Yara N L F de Maria
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| | - David Aciole Barbosa
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| | - Fabiano B Menegidio
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| | | | | | - Valquíria C Alencar
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Brazil
| | - Juliana F S Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Brazil
| | | | - Miguel L Batista
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
- Department of Biochemistry, Boston University School of Medicine, USA
| | - Luiz R Nunes
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), Brazil
| | - Daniela L Jabes
- Núcleo Integrado de Biotecnologia, Universidade de Mogi das Cruzes (UMC), Brazil
| |
Collapse
|
7
|
Adiliaghdam F, Cavallaro P, Mohad V, Almpani M, Kühn F, Gharedaghi MH, Najibi M, Rahme LG, Hodin RA. Targeting the gut to prevent sepsis from a cutaneous burn. JCI Insight 2020; 5:137128. [PMID: 33004693 PMCID: PMC7566703 DOI: 10.1172/jci.insight.137128] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Severe burn injury induces gut barrier dysfunction and subsequently a profound systemic inflammatory response. In the present study, we examined the role of the small intestinal brush border enzyme, intestinal alkaline phosphatase (IAP), in preserving gut barrier function and preventing systemic inflammation after burn wound infection in mice. Mice were subjected to a 30% total body surface area dorsal burn with or without intradermal injection of Pseudomonas aeruginosa. Mice were gavaged with 2000 units of IAP or vehicle at 3 and 12 hours after the insult. We found that both endogenously produced and exogenously supplemented IAP significantly reduced gut barrier damage, decreased bacterial translocation to the systemic organs, attenuated systemic inflammation, and improved survival in this burn wound infection model. IAP attenuated liver inflammation and reduced the proinflammatory characteristics of portal serum. Furthermore, we found that intestinal luminal contents of burn wound-infected mice negatively impacted the intestinal epithelial integrity compared with luminal contents of control mice and that IAP supplementation preserved monolayer integrity. These results indicate that oral IAP therapy may represent an approach to preserving gut barrier function, blocking proinflammatory triggers from entering the portal system, preventing gut-induced systemic inflammation, and improving survival after severe burn injuries.
Collapse
Affiliation(s)
- Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul Cavallaro
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vidisha Mohad
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Almpani
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian Kühn
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of General, Visceral and Transplant Surgery, Hospital of the University of Munich, Munich, Germany
| | - Mohammad Hadi Gharedaghi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mehran Najibi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laurence G. Rahme
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Urban RJ, Pyles RB, Stewart CJ, Ajami N, Randolph KM, Durham WJ, Danesi CP, Dillon EL, Summons JR, Singh CK, Morrison M, Kreber LA, Masel B, Miller AL, Wright TJ, Sheffield-Moore M. Altered Fecal Microbiome Years after Traumatic Brain Injury. J Neurotrauma 2020; 37:1037-1051. [DOI: 10.1089/neu.2019.6688] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Randall J. Urban
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
| | - Richard B. Pyles
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas
| | - Christopher J. Stewart
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas
| | - Nadim Ajami
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas
| | - Kathleen M. Randolph
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
- Department of Health and Kinesiology, Texas A&M University, College Station, Texas
| | - William J. Durham
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
| | - Christopher P. Danesi
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
| | - E. Lichar Dillon
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
| | | | | | | | | | - Brent Masel
- Centre for Neuroskills, Bakersfield, California
- Department of Neurology, The University of Texas Medical Branch, Galveston, Texas
- Brain Injury Association of America, Vienna, Virginia
| | - Aaron L. Miller
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, Texas
| | - Traver J. Wright
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
- Department of Health and Kinesiology, Texas A&M University, College Station, Texas
| | - Melinda Sheffield-Moore
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas
- Department of Health and Kinesiology, Texas A&M University, College Station, Texas
| |
Collapse
|
9
|
Ke J, Bian X, Liu H, Li B, Huo R. Edaravone reduces oxidative stress and intestinal cell apoptosis after burn through up-regulating miR-320 expression. Mol Med 2019; 25:54. [PMID: 31829167 PMCID: PMC6907153 DOI: 10.1186/s10020-019-0122-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Intestinal mucosa barrier dysfunction after burn injury is an important factor for causing mortality of burn patients. The current study established a burn model in rats and used a free radical scavenger edaravone (ED) to treat the rats, so as to investigate the effect of edaravone on intestinal mucosa barrier after burn injury. METHODS Anesthetized rats were subjected to 40% total body surface area water burn immediately, followed by treatment with ED, scrambled antagomir, or antagomiR-320. Intestinal mucosa damage was observed by hematoxylin-eosin staining and graded by colon mucosal damage index (CMDI) score. The contents of total sulfhydryl (TSH), superoxide dismutase (SOD), catalase (CAT) and malondialdehyde (MDA) were determined by spectrophotometry. Cell apoptosis, protein relative expression,and the in situ expressions of p-Akt and p-Bad were detected by flow cytometry, Western blotting and immunohistochemistry, respectively. The miR-320 expression was determined by quantitative real-time polymerase chain reaction. RESULTS ED alleviated intestinal mucosal damage caused by burn injury, down-regulated the levels of MDA, cytochrome C, cleaved caspase-9 and cleaved caspase-3, but up-regulated the levels of TSH, SOD, CAT and Bcl-2. We also found that ED could reduce oxidative stress, inhibit cell apoptosis, increase the expressions of p-Akt, p-Bad and miR-320, and decrease PTEN expression. PTEN was predicted to be the target gene for miR-320, and cell apoptosis could be promoted by inhibiting miR-320 expression. CONCLUSION ED regulates Akt/Bad/Caspase signaling cascade to reduce apoptosis and oxidative stress through up-regulating miR-320 expression and down-regulating PTEN expression, thus protecting the intestinal mucosal barrier of rats from burn injury.
Collapse
Affiliation(s)
- Jiaxiang Ke
- Burn and Plastic Section, Qingdao Municipal Hospital Affiliated to Shandong University, Qingdao, China
| | - Xi Bian
- Burn and Plastic Section, Qingdao Municipal Hospital Affiliated to Shandong University, Qingdao, China
| | - Hu Liu
- Burn and Plastic Section, Qingdao Municipal Hospital Affiliated to Shandong University, Qingdao, China
| | - Bei Li
- Burn and Plastic Section, Qingdao Municipal Hospital Affiliated to Shandong University, Qingdao, China
| | - Ran Huo
- Burn and Plastic Section, Shandong Province Hospital Affiliated to Shandong University, Jiaozhou Road, Shibei District, Qingdao, 266011, Shandong Province, China.
| |
Collapse
|
10
|
Hashemi FB, Behrouz B, Irajian G, Laghaei P, Korpi F, Fatemi MJ. A trivalent vaccine consisting of "flagellin A+B and pilin" protects against Pseudomonas aeruginosa infection in a murine burn model. Microb Pathog 2019; 138:103697. [PMID: 31465785 DOI: 10.1016/j.micpath.2019.103697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/22/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly achieves antibiotic resistance, and thus, developing an effective vaccine is critically important for combating P. aeruginosa infection. Flagella and pili play important roles in colonization of P. aeruginosa at the burn wound site and its subsequent dissemination to deeper tissue and organs. In the present study, we evaluated protective efficacy of a trivalent vaccine containing flagellins A and B (FlaA + FlaB) + pilin (PilA) in a murine burn model of infection. "FlaA + FlaB + PilA" induced greater protection in P. aeruginosa murine burn model than the single components alone, and it showed broad immune protection against P. aeruginosa strains. Immunization with "FlaA + FlaB + PilA" induced strong opsonophagocytic antibodies and resulted in reduced bacterial loads, systemic IL-12/IL-10 cytokine expression, and increased survival after challenge with three times lethal dose fifty (LD50) of P. eruginosa strains. Moreover, the protective efficacy of "FlaA + FlaB + PilA" vaccination was largely attributed to specific antibodies. Taken together, these data further confirm that the protective effects of "FlaA + FlaB + PilA" vaccine significantly enhance efficacy compared with antibodies against either mono or divalent antigen, and that the former broadens the coverage against P. eruginosa strains that express two of the three antigens.
Collapse
Affiliation(s)
- Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Laghaei
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Korpi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Abstract
Humans are a colonized with trillions of commensal microorganisms which exert a profound effect on normal host physiology and immune function through an abundance of genetic and metabolic by-products. Although the commensal microbiome has beneficial functions to host physiology, perturbations of the composition of the commensal microbiome or the homeostatic mucosal environment can lead to the induction of immune pathology and systemic inflammation. In the context of cancer progression or response to immune therapy, this inflammation can be detrimental, resulting in tumor growth and the promotion of immune suppression. On the other hand, significant associations have been identified whereby certain commensal microorganisms are able to enhance T cell function or are required for tumor control in cancer patients treated with certain immune therapies and chemotherapies. The focus of this chapter is to highlight the role of the commensal microbiome during tumor progression and in response to immune therapies.
Collapse
|
12
|
INDICATORS CELL CYCLE AND DNA FRAGMENTATION IN CELLS OF SMALL INTESTINE MUCOSA 14, 21 AND 30 DAYS AFTER SKIN BURNS ON THE BACKGROUND OF PRE-LIMINARY INFUSION OF SOLUTION LACTOPROTEIN WITH SORBITOL OR HAES-LX 5%. WORLD OF MEDICINE AND BIOLOGY 2018. [DOI: 10.26724/2079-8334-2017-4-62-104-108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
13
|
Ji LJ, Shi J, Lu JM, Huang QM. MiR-150 alleviates neuropathic pain via inhibiting toll-like receptor 5. J Cell Biochem 2017; 119:1017-1026. [PMID: 28685867 DOI: 10.1002/jcb.26269] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/06/2017] [Indexed: 12/30/2022]
Abstract
MicroRNAs (miRNAs) are reported as vital participators in the pathophysiological course of neuropathic pain. However, the underlying mechanisms of the functional roles of miRNAs in neuropathic pain are largely unknown. This study was designed to explore the potential role of miR-150 in regulating the process of neuropathic pain in a rat model established by chronic sciatic nerve injury (CCI). Overexpression of miR-150 greatly alleviated neuropathic pain development and reduced inflammatory cytokine expression, including COX-2, interleukin IL-6, and tumor necrosis factor (TNF)-α in CCI rats. By bioinformatic analysis, 3'-untranslated region (UTR) of Toll-like receptor (TLR5) was predicted to be a target of miR-150. TLR5 commonly serves as an important regulator of inflammation. Overexpression of miR-150 significantly suppressed the expression of TLR5 in vitro and in vivo. Furthermore, upregulation of TLR5 decreased the miR-150 expression and downregulation of TLR5 increased miR-150, respectively. Overexpression of TLR5 significantly reversed the miR-150-induced suppressive effects on neuropathic pain. In conclusion, our current study indicates that miR-150 may inhibit neuropathic pain development of CCI rats through inhibiting TLR5-mediated neuroinflammation. Our findings suggest that miR-150 may provide a novel therapeutic target for neuropathic pain treatment.
Collapse
Affiliation(s)
- Li-Juan Ji
- Department of Sport Medicine and Rehabilitation Center, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jing Shi
- Geriatric Department,The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Min Lu
- Department of Neurology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Qiang-Min Huang
- Department of Sport Medicine and Rehabilitation Center, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
14
|
The influence of the commensal microbiota on distal tumor-promoting inflammation. Semin Immunol 2017; 32:62-73. [PMID: 28687194 DOI: 10.1016/j.smim.2017.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023]
Abstract
Commensal microbes inhabit barrier surfaces, providing a first line of defense against invading pathogens, aiding in metabolic function of the host, and playing a vital role in immune development and function. Several recent studies have demonstrated that commensal microbes influence systemic immune function and homeostasis. For patients with extramucosal cancers, or cancers occurring distal to barrier surfaces, the role of commensal microbes in influencing tumor progression is beginning to be appreciated. Extrinsic factors such as chronic inflammation, antibiotics, and chemotherapy dysregulate commensal homeostasis and drive tumor-promoting systemic inflammation through a variety of mechanisms, including disruption of barrier function and bacterial translocation, release of soluble inflammatory mediators, and systemic changes in metabolic output. Conversely, it has also been demonstrated that certain immune therapies, immunogenic chemotherapies, and checkpoint inhibitors rely on the commensal microbiota to facilitate anti-tumor immune responses. Thus, it is evident that the mechanisms associated with commensal microbe facilitation of both pro- and anti-tumor immune responses are context dependent and rely upon a variety of factors present within the tumor microenvironment and systemic periphery. The goal of this review is to highlight the various contexts during which commensal microbes orchestrate systemic immune function with a focus on describing possible scenarios where the loss of microbial homeostasis enhances tumor progression.
Collapse
|
15
|
Correction: Intraluminal Flagellin Differentially Contributes to Gut Dysbiosis and Systemic Inflammation following Burn Injury. PLoS One 2017; 12:e0170722. [PMID: 28103320 PMCID: PMC5245817 DOI: 10.1371/journal.pone.0170722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|