1
|
Nataraj NM, Sillas RG, Herrmann BI, Shin S, Brodsky IE. Blockade of IKK signaling induces RIPK1-independent apoptosis in human macrophages. PLoS Pathog 2024; 20:e1012469. [PMID: 39186805 PMCID: PMC11407650 DOI: 10.1371/journal.ppat.1012469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/17/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
Regulated cell death in response to microbial infection plays an important role in immune defense and is triggered by pathogen disruption of essential cellular pathways. Gram-negative bacterial pathogens in the Yersinia genus disrupt NF-κB signaling via translocated effectors injected by a type III secretion system, thereby preventing induction of cytokine production and antimicrobial defense. In murine models of infection, Yersinia blockade of NF-κB signaling triggers cell-extrinsic apoptosis through Receptor Interacting Serine-Threonine Protein Kinase 1 (RIPK1) and caspase-8, which is required for bacterial clearance and host survival. Unexpectedly, we find that human macrophages undergo apoptosis independently of RIPK1 in response to Yersinia or chemical blockade of IKKβ. Instead, IKK blockade led to decreased cFLIP expression, and overexpression of cFLIP contributed to protection from IKK blockade-induced apoptosis in human macrophages. We found that IKK blockade also induces RIPK1 kinase activity-independent apoptosis in human T cells and human pancreatic cells. Altogether, our data indicate that, in contrast to murine cells, blockade of IKK activity in human cells triggers a distinct apoptosis pathway that is independent of RIPK1 kinase activity. These findings have implications for the contribution of RIPK1 to cell death in human cells and the efficacy of RIPK1 inhibition in human diseases.
Collapse
Affiliation(s)
- Neha M Nataraj
- Institute for Immunology & Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Reyna Garcia Sillas
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Beatrice I Herrmann
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Sunny Shin
- Institute for Immunology & Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Igor E Brodsky
- Institute for Immunology & Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
2
|
Simón-Fuentes M, Ríos I, Herrero C, Lasala F, Labiod N, Luczkowiak J, Roy-Vallejo E, Fernández de Córdoba-Oñate S, Delgado-Wicke P, Bustos M, Fernández-Ruiz E, Colmenares M, Puig-Kröger A, Delgado R, Vega MA, Corbí ÁL, Domínguez-Soto Á. MAFB shapes human monocyte-derived macrophage response to SARS-CoV-2 and controls severe COVID-19 biomarker expression. JCI Insight 2023; 8:e172862. [PMID: 37917179 PMCID: PMC10807725 DOI: 10.1172/jci.insight.172862] [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: 06/09/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
Monocyte-derived macrophages, the major source of pathogenic macrophages in COVID-19, are oppositely instructed by macrophage CSF (M-CSF) or granulocyte macrophage CSF (GM-CSF), which promote the generation of antiinflammatory/immunosuppressive MAFB+ (M-MØ) or proinflammatory macrophages (GM-MØ), respectively. The transcriptional profile of prevailing macrophage subsets in severe COVID-19 led us to hypothesize that MAFB shapes the transcriptome of pulmonary macrophages driving severe COVID-19 pathogenesis. We have now assessed the role of MAFB in the response of monocyte-derived macrophages to SARS-CoV-2 through genetic and pharmacological approaches, and we demonstrate that MAFB regulated the expression of the genes that define pulmonary pathogenic macrophages in severe COVID-19. Indeed, SARS-CoV-2 potentiated the expression of MAFB and MAFB-regulated genes in M-MØ and GM-MØ, where MAFB upregulated the expression of profibrotic and neutrophil-attracting factors. Thus, MAFB determines the transcriptome and functions of the monocyte-derived macrophage subsets that underlie pulmonary pathogenesis in severe COVID-19 and controls the expression of potentially useful biomarkers for COVID-19 severity.
Collapse
Affiliation(s)
- Miriam Simón-Fuentes
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Israel Ríos
- Immunometabolism and Inflammation Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Cristina Herrero
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Fátima Lasala
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Nuria Labiod
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Emilia Roy-Vallejo
- Rheumatology Department, University Hospital La Princesa and Research Institute, Madrid, Spain
| | | | - Pablo Delgado-Wicke
- Molecular Biology Unit, University Hospital La Princesa and Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - Matilde Bustos
- Institute of Biomedicine of Seville (IBiS), Spanish National Research Council (CSIC), University of Seville, Virgen del Rocio University Hospital (HUVR), Seville, Spain
| | - Elena Fernández-Ruiz
- Molecular Biology Unit, University Hospital La Princesa and Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Colmenares
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Amaya Puig-Kröger
- Immunometabolism and Inflammation Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Miguel A. Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | |
Collapse
|
3
|
Jones RP. Addressing the Knowledge Deficit in Hospital Bed Planning and Defining an Optimum Region for the Number of Different Types of Hospital Beds in an Effective Health Care System. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:7171. [PMID: 38131722 PMCID: PMC11080941 DOI: 10.3390/ijerph20247171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Based upon 30-years of research by the author, a new approach to hospital bed planning and international benchmarking is proposed. The number of hospital beds per 1000 people is commonly used to compare international bed numbers. This method is flawed because it does not consider population age structure or the effect of nearness-to-death on hospital utilization. Deaths are also serving as a proxy for wider bed demand arising from undetected outbreaks of 3000 species of human pathogens. To remedy this problem, a new approach to bed modeling has been developed that plots beds per 1000 deaths against deaths per 1000 population. Lines of equivalence can be drawn on the plot to delineate countries with a higher or lower bed supply. This method is extended to attempt to define the optimum region for bed supply in an effective health care system. England is used as an example of a health system descending into operational chaos due to too few beds and manpower. The former Soviet bloc countries represent a health system overly dependent on hospital beds. Several countries also show evidence of overutilization of hospital beds. The new method is used to define a potential range for bed supply and manpower where the most effective health systems currently reside. The method is applied to total curative beds, medical beds, psychiatric beds, critical care, geriatric care, etc., and can also be used to compare different types of healthcare staff, i.e., nurses, physicians, and surgeons. Issues surrounding the optimum hospital size and the optimum average occupancy will also be discussed. The role of poor policy in the English NHS is used to show how the NHS has been led into a bed crisis. The method is also extended beyond international benchmarking to illustrate how it can be applied at a local or regional level in the process of long-term bed planning. Issues regarding the volatility in hospital admissions are also addressed to explain the need for surge capacity and why an adequate average bed occupancy margin is required for an optimally functioning hospital.
Collapse
|
4
|
Kushwaha A, Agarwal V. Pseudomonas aeruginosa quorum-sensing molecule N-(3-oxododecanoyl)-L-homoserine lactone mediates Ca +2 dysregulation, mitochondrial dysfunction, and apoptosis in human peripheral blood lymphocytes. Heliyon 2023; 9:e21462. [PMID: 38027911 PMCID: PMC10660034 DOI: 10.1016/j.heliyon.2023.e21462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/01/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023] Open
Abstract
N-(3-oxododecanoyl)-l-homoserine lactone is a Pseudomonas aeruginosa secreted quorum-sensing molecule that mediates the secretion of virulence factors, biofilm formation and plays a pivotal role in proliferation and persistence in the host. Apart from regulating quorum-sensing, the autoinducer signal molecule N-(3-oxododecanoyl)-l-homoserine lactone (3O-C12-HSL or C12) of a LasI-LasR circuit exhibits immunomodulatory effects and induces apoptosis in various host cells. However, the precise pathophysiological impact of C12 on human peripheral blood lymphocytes and its involvement in mitochondrial dysfunction remained largely elusive. In this study, the results suggest that C12 (100 μM) induces upregulation of cytosolic and mitochondrial Ca+2 levels and triggers mitochondrial dysfunction through the generation of mitochondrial ROS (mROS), disruption of mitochondrial transmembrane potential (ΔΨm), and opening of the mitochondrial permeability transition pore (mPTP). Additionally, it was observed that C12 induces phosphatidylserine (PS) exposure and promotes apoptosis in human peripheral blood lymphocytes. However, apoptosis plays a critical role in the homeostasis and development of lymphocytes, whereas enhanced apoptosis can cause immunodeficiency through cell loss. These findings suggest that C12 exerts a detrimental effect on lymphocytes by mediating mitochondrial dysfunction and enhancing apoptosis, which might further impair the effective mounting of immune responses during Pseudomonas aeruginosa-associated infections.
Collapse
Affiliation(s)
- Ankit Kushwaha
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| | - Vishnu Agarwal
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| |
Collapse
|
5
|
Scott TJ, Larsen TJ, Brock DA, Uhm SYS, Queller DC, Strassmann JE. Symbiotic bacteria, immune-like sentinel cells, and the response to pathogens in a social amoeba. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230727. [PMID: 37593719 PMCID: PMC10427822 DOI: 10.1098/rsos.230727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023]
Abstract
Some endosymbionts living within a host must modulate their hosts' immune systems in order to infect and persist. We studied the effect of a bacterial endosymbiont on a facultatively multicellular social amoeba host. Aggregates of the amoeba Dictyostelium discoideum contain a subpopulation of sentinel cells that function akin to the immune systems of more conventional multicellular organisms. Sentinel cells sequester and discard toxins from D. discoideum aggregates and may play a central role in defence against pathogens. We measured the number and functionality of sentinel cells in aggregates of D. discoideum infected by bacterial endosymbionts in the genus Paraburkholderia. Infected D. discoideum produced fewer and less functional sentinel cells, suggesting that Paraburkholderia may interfere with its host's immune system. Despite impaired sentinel cells, however, infected D. discoideum were less sensitive to ethidium bromide toxicity, suggesting that Paraburkholderia may also have a protective effect on its host. By contrast, D. discoideum infected by Paraburkholderia did not show differences in their sensitivity to two non-symbiotic pathogens. Our results expand previous work on yet another aspect of the complicated relationship between D. discoideum and Paraburkholderia, which has considerable potential as a model for the study of symbiosis.
Collapse
Affiliation(s)
- Trey J. Scott
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Tyler J. Larsen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Debra A. Brock
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - So Yeon Stacey Uhm
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - David C. Queller
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Joan E. Strassmann
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
6
|
Zhu XY, Wang ML, Cai M, Nan XM, Zhao YG, Xiong BH, Yang L. Protein Expression Profiles in Exosomes of Bovine Mammary Epithelial Cell Line MAC-T Infected with Staphylococcus aureus. Appl Environ Microbiol 2023; 89:e0174322. [PMID: 36939340 PMCID: PMC10132110 DOI: 10.1128/aem.01743-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/20/2023] [Indexed: 03/21/2023] Open
Abstract
Mastitis is a common and widespread infectious disease in dairy farms around the world, resulting in reduced milk production and quality. Staphylococcus aureus is one of the main pathogenic bacteria causing subclinical mastitis in dairy cows. S. aureus can activate inflammatory signaling pathways in bovine mammary epithelial cells. Exosomes produced by cells can directly transfer pathogen-related molecules from cell to cell, thus affecting the process of infection. Protein is the material basis of the immune defense function in the body; therefore, a comprehensive comparison of proteins in exosomes derived from S. aureus-infected (SA group) and normal (control group [C group]) bovine mammary epithelial MAC-T cells was performed using shotgun proteomics by a DIA approach. A total of 7,070 proteins were identified and quantified. Compared with the C group, there were 802 differentially expressed proteins (DEPs) identified in the SA group (absolute log2 fold change [|log2FC|] of ≥0.58; false discovery rate [FDR] of <0.05), among which 325 proteins were upregulated and 477 were downregulated. The upregulated proteins, including complement 3 (C3), integrin alpha-6 (ITGA6), apolipoprotein A1 (APOA1), annexin A2 (ANXA2), tripeptidyl peptidase II (TPP2), keratin 8 (KRT8), and recombinant desmoyokin (AHNAK), are involved mostly in host defense against pathogens, inflammation, and cell structure maintenance. KEGG enrichment analysis indicated that DEPs in S. aureus infection were involved in the complement and coagulation cascade, phagosome, extracellular matrix (ECM)-receptor interaction, and focal adhesion pathways. The results of this study provide novel information about proteins in the exosomes of MAC-T cells infected with S. aureus and could contribute to an understanding of the infectious mechanism of bovine mastitis. IMPORTANCE Mastitis is a widespread infectious disease in dairy farms, resulting in reduced milk production and quality. Staphylococcus aureus is one of the main pathogenic bacteria causing subclinical mastitis. Exosomes contain proteins, lipids, and nucleic acids, which are involved in many physiological and pathological functions. The expression of proteins in exosomes derived from bovine mammary epithelial cells infected by S. aureus is still barely understood. These results provide novel information about MAC-T-derived exosomal proteins, reveal insights into their functions, and lay a foundation for further studying the biological function of exosomes during the inflammatory response.
Collapse
Affiliation(s)
- Xiao-Yan Zhu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Meng-Ling Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Meng Cai
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xue-Mei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yi-Guang Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ben-Hai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liang Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
7
|
Nath SK, Pankajakshan P, Sharma T, Kumari P, Shinde S, Garg N, Mathur K, Arambam N, Harjani D, Raj M, Kwatra G, Venkatesh S, Choudhoury A, Bano S, Tayal P, Sharan M, Arora R, Strych U, Hotez PJ, Bottazzi ME, Rawal K. A Data-Driven Approach to Construct a Molecular Map of Trypanosoma cruzi to Identify Drugs and Vaccine Targets. Vaccines (Basel) 2023; 11:vaccines11020267. [PMID: 36851145 PMCID: PMC9963959 DOI: 10.3390/vaccines11020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Chagas disease (CD) is endemic in large parts of Central and South America, as well as in Texas and the southern regions of the United States. Successful parasites, such as the causative agent of CD, Trypanosoma cruzi have adapted to specific hosts during their phylogenesis. In this work, we have assembled an interactive network of the complex relations that occur between molecules within T. cruzi. An expert curation strategy was combined with a text-mining approach to screen 10,234 full-length research articles and over 200,000 abstracts relevant to T. cruzi. We obtained a scale-free network consisting of 1055 nodes and 874 edges, and composed of 838 proteins, 43 genes, 20 complexes, 9 RNAs, 36 simple molecules, 81 phenotypes, and 37 known pharmaceuticals. Further, we deployed an automated docking pipeline to conduct large-scale docking studies involving several thousand drugs and potential targets to identify network-based binding propensities. These experiments have revealed that the existing FDA-approved drugs benznidazole (Bz) and nifurtimox (Nf) show comparatively high binding energies to the T. cruzi network proteins (e.g., PIF1 helicase-like protein, trans-sialidase), when compared with control datasets consisting of proteins from other pathogens. We envisage this work to be of value to those interested in finding new vaccines for CD, as well as drugs against the T. cruzi parasite.
Collapse
Affiliation(s)
- Swarsat Kaushik Nath
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Preeti Pankajakshan
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Trapti Sharma
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Priya Kumari
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Sweety Shinde
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Nikita Garg
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Kartavya Mathur
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Nevidita Arambam
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Divyank Harjani
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Manpriya Raj
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Garwit Kwatra
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Sayantan Venkatesh
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Alakto Choudhoury
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Saima Bano
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Prashansa Tayal
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Mahek Sharan
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Ruchika Arora
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Kamal Rawal
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
- Correspondence:
| |
Collapse
|
8
|
Zhang J, Zhang S, Sun X, Xu X. Comparative transcriptome analysis reveals the immune response of turbot (Scophthalmus maximus) induced by inactivated bivalent vaccine. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108461. [PMID: 36462744 DOI: 10.1016/j.fsi.2022.108461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Vibrio species are important pathogens that affect a wide range of farmed fish. Vaccination is regarded as the most efficacious strategy for fighting bacterial infections. However, the underlying mechanisms remain to be elucidated. In the present study, a comparative transcriptome analysis was performed on the spleens from turbot (Scophthalmus maximus) induced by an inactivated bivalent vaccine (Vibrio anguillarum and Vibrio harveyi, IVVah1) at 4 week and 1 day post further challenge. Strong immune responses were induced by the bivalent vaccine, besides differentially expressed genes (DEGs) associated with adaptive immunity, more innate immunity-related DEGs were detected. At the late stage of vaccination, immune-related molecules associated with pattern recognition receptors, inflammatory factors, complement and coagulation cascade-related components, and antigen processing and presentation were significantly regulated, and some of them were even further up-regulated after the bacterial challenge, indicating the cooperation of multiple immune processes during the vaccine immunization process. In addition to the terms or pathways associated with the immune response, enrichment analysis revealed multiple significantly enriched terms/pathways associated with the response to stimulus/stress, homeostasis, metabolism, and biosynthesis, suggesting that a defensive status was established by the bivalent vaccine. This study furnishes new insights into the internal mechanism of immunity upon a combined vaccine administrating in turbot and lays a foundation for developing highly immunogenic vaccines in teleost.
Collapse
Affiliation(s)
- Jian Zhang
- School of Ocean, Yantai University, Yantai, China
| | | | - Xiangyi Sun
- School of Ocean, Yantai University, Yantai, China
| | - Xiudan Xu
- School of Ocean, Yantai University, Yantai, China.
| |
Collapse
|
9
|
Lin Y, Mi D, Hou Y, Lin M, Xie Q, Niu X, Chen Y, He C, Tao J, Li C. Systematic analysis of the roles of c-di-GMP signaling in Xanthomonas oryzae pv. oryzae virulence. FEMS Microbiol Lett 2022; 369:6650349. [PMID: 35883214 DOI: 10.1093/femsle/fnac068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/09/2022] [Accepted: 07/22/2022] [Indexed: 11/14/2022] Open
Abstract
Cyclic di-guanosine monophosphate (c-di-GMP) is a ubiquitous second messenger that is essential to bacterial adaptation to environments. Cellular c-di-GMP level is regulated by the diguanylate cyclases and the phosphodiesterases, and the signal transduction depends on its receptors. In Xanthomonas oryzae pv. oryzae strain PXO99A, 37 genes were predicted to encode GGDEF, EAL, GGDEF/EAL, HD-GYP, FleQ, MshE, PilZ, CuxR, Clp, YajQ proteins that may be involved in c-di-GMP turnover or function as c-di-GMP receptors. Although the functions of some of these genes have been studied, but the rest have not been extensively studied. Here, we deleted these 37 genes from PXO99A and analyzed the virulence, motility, biofilm and EPS production of these mutants. Our results show that most of these genes are required for PXO99A virulence, motility, biofilm formation or exopolysaccharide production. Although some of them have been reported in previous studies, we found four novel genes (gedpX8, gdpX11, pliZX4 and yajQ) are implicated in X. oryzae pv. oryzae virulence. Our data demonstrate that c-di-GMP signaling is vital for X. oryzae pv. oryzae virulence and some virulence-related factors production, but there is no positive correlation between them in most cases. Taken together, our systematic research provides a new light to understand the c-di-GMP signaling network in X. oryzae pv. oryzae.
Collapse
Affiliation(s)
- Yunuan Lin
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,School of Life Sciences
| | - Duo Mi
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,College of Tropical Crops, Hainan University, Haikou 570228, China
| | - Yunyu Hou
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,School of Life Sciences
| | - Maojuan Lin
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,College of Tropical Crops, Hainan University, Haikou 570228, China
| | - Qingbiao Xie
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,School of Life Sciences
| | - Xiaolei Niu
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,College of Tropical Crops, Hainan University, Haikou 570228, China
| | - Yinhua Chen
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,School of Life Sciences
| | - Chaozu He
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,College of Tropical Crops, Hainan University, Haikou 570228, China
| | - Jun Tao
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,College of Tropical Crops, Hainan University, Haikou 570228, China
| | - Chunxia Li
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresources.,College of Tropical Crops, Hainan University, Haikou 570228, China
| |
Collapse
|
10
|
Ventura-Santana E, Ninan JR, Snyder CM, Okeke EB. Neutrophil Extracellular Traps, Sepsis and COVID-19 - A Tripod Stand. Front Immunol 2022; 13:902206. [PMID: 35757734 PMCID: PMC9226304 DOI: 10.3389/fimmu.2022.902206] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the current coronavirus disease 2019 (COVID-19) pandemic. Majority of COVID-19 patients have mild disease but about 20% of COVID-19 patients progress to severe disease. These patients end up in the intensive care unit (ICU) with clinical manifestations of acute respiratory distress syndrome (ARDS) and sepsis. The formation of neutrophil extracellular traps (NETs) has also been associated with severe COVID-19. Understanding of the immunopathology of COVID-19 is critical for the development of effective therapeutics. In this article, we discuss evidence indicating that severe COVID-19 has clinical presentations consistent with the definitions of viral sepsis. We highlight the role of neutrophils and NETs formation in the pathogenesis of severe COVID-19. Finally, we highlight the potential of therapies inhibiting NETs formation for the treatment of COVID-19.
Collapse
Affiliation(s)
- Esmeiry Ventura-Santana
- Department of Biology, State University of New York at Fredonia, Fredonia, NY, United States
| | - Joshua R Ninan
- Department of Biology, State University of New York at Fredonia, Fredonia, NY, United States
| | - Caitlin M Snyder
- Department of Biology, State University of New York at Fredonia, Fredonia, NY, United States
| | - Emeka B Okeke
- Department of Biology, State University of New York at Fredonia, Fredonia, NY, United States
| |
Collapse
|
11
|
Kufer TA, Kaparakis-Liaskos M. A Brief Introduction to Effector-Triggered Immunity. Methods Mol Biol 2022; 2523:1-8. [PMID: 35759187 DOI: 10.1007/978-1-0716-2449-4_1] [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] [Indexed: 06/15/2023]
Abstract
Detection of microbes by the host is essential to restrict microbial colonization, to clear pathogens, and to mount adapted defense reactions, and thus is the key function of the innate immune systems of plants and mammals. Here we provide an introduction into pathogen recognition by the innate immune system of both plants and animals. We will particularly focus on the concept of effector-triggered immunity, and similarities and differences in its function between plants and animals.
Collapse
Affiliation(s)
- Thomas A Kufer
- Department of Immunology, Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Maria Kaparakis-Liaskos
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
- Research Centre for Extracellular Vesicles, School of Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Abstract
Pathogens have evolved smart strategies to invade hosts and hijack their immune responses. One such strategy is the targeting of the host RhoGTPases by toxins or virulence factors to hijack the cytoskeleton dynamic and immune processes. In response to this microbial attack, the host has evolved an elegant strategy to monitor the function of virulence factors and toxins by sensing the abnormal activity of RhoGTPases. This innate immune strategy of sensing bacterial effector targeting RhoGTPase appears to be a bona fide example of effector-triggered immunity (ETI). Here, we review recently discovered mechanisms by which the host can sense the activity of these toxins through NOD and NOD-like receptors (NLRs).
Collapse
Affiliation(s)
| | - Laurent Boyer
- Université Côte d’Azur, Inserm, C3M, Nice, France
- * E-mail:
| |
Collapse
|
13
|
Palau M, Piqué N, Ramírez-Lázaro MJ, Lario S, Calvet X, Miñana-Galbis D. Whole-Genome Sequencing and Comparative Genomics of Three Helicobacter pylori Strains Isolated from the Stomach of a Patient with Adenocarcinoma. Pathogens 2021; 10:331. [PMID: 33809022 PMCID: PMC7998635 DOI: 10.3390/pathogens10030331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 12/28/2022] Open
Abstract
Helicobacter pylori is a common pathogen associated with several severe digestive diseases. Although multiple virulence factors have been described, it is still unclear the role of virulence factors on H. pylori pathogenesis and disease progression. Whole genome sequencing could help to find genetic markers of virulence strains. In this work, we analyzed three complete genomes from isolates obtained at the same point in time from a stomach of a patient with adenocarcinoma, using multiple available bioinformatics tools. The genome analysis of the strains B508A-S1, B508A-T2A and B508A-T4 revealed that they were cagA, babA and sabB/hopO negative. The differences among the three genomes were mainly related to outer membrane proteins, methylases, restriction modification systems and flagellar biosynthesis proteins. The strain B508A-T2A was the only one presenting the genotype vacA s1, and had the most distinct genome as it exhibited fewer shared genes, higher number of unique genes, and more polymorphisms were found in this genome. With all the accumulated information, no significant differences were found among the isolates regarding virulence and origin of the isolates. Nevertheless, some B508A-T2A genome characteristics could be linked to the pathogenicity of H. pylori.
Collapse
Affiliation(s)
- Montserrat Palau
- Secció de Microbiologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Catalonia, Spain; (M.P.); (N.P.)
| | - Núria Piqué
- Secció de Microbiologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Catalonia, Spain; (M.P.); (N.P.)
| | - M. José Ramírez-Lázaro
- Digestive Diseases Service, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Parc Tauli 1, 08208 Sabadell, Catalonia, Spain; (M.J.R.-L.); (S.L.); (X.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Monforte de Lemos 3–5, 28029 Madrid, Community of Madrid, Spain
| | - Sergio Lario
- Digestive Diseases Service, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Parc Tauli 1, 08208 Sabadell, Catalonia, Spain; (M.J.R.-L.); (S.L.); (X.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Monforte de Lemos 3–5, 28029 Madrid, Community of Madrid, Spain
| | - Xavier Calvet
- Digestive Diseases Service, Hospital de Sabadell, Institut Universitari Parc Taulí-UAB, Parc Tauli 1, 08208 Sabadell, Catalonia, Spain; (M.J.R.-L.); (S.L.); (X.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Monforte de Lemos 3–5, 28029 Madrid, Community of Madrid, Spain
| | - David Miñana-Galbis
- Secció de Microbiologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Catalonia, Spain; (M.P.); (N.P.)
| |
Collapse
|
14
|
Ngwaga T, Chauhan D, Shames SR. Mechanisms of Effector-Mediated Immunity Revealed by the Accidental Human Pathogen Legionella pneumophila. Front Cell Infect Microbiol 2021; 10:593823. [PMID: 33614523 PMCID: PMC7886983 DOI: 10.3389/fcimb.2020.593823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022] Open
Abstract
Many Gram-negative bacterial pathogens employ translocated virulence factors, termed effector proteins, to facilitate their parasitism of host cells and evade host anti-microbial defenses. However, eukaryotes have evolved to detect effector-mediated virulence strategies through a phenomenon termed effector-triggered immunity (ETI). Although ETI was discovered in plants, a growing body of literature demonstrates that metazoans also utilize effector-mediated immunity to detect and clear bacterial pathogens. This mini review is focused on mechanisms of effector-mediated immune responses by the accidental human pathogen Legionella pneumophila. We highlight recent advancements in the field and discuss the future prospects of harnessing effectors for the development of novel therapeutics, a critical need due to the prevalence and rapid spread of antibiotic resistance.
Collapse
Affiliation(s)
- Tshegofatso Ngwaga
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Deepika Chauhan
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Stephanie R Shames
- Division of Biology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
15
|
Matos ADO, Dantas PHDS, Silva-Sales M, Sales-Campos H. TREM-1 isoforms in bacterial infections: to immune modulation and beyond. Crit Rev Microbiol 2021; 47:290-306. [PMID: 33522328 DOI: 10.1080/1040841x.2021.1878106] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The triggering receptor expressed on myeloid cells 1 (TREM-1) is an innate immunity receptor associated with the amplification of inflammation in sterile and non-sterile inflammatory disorders. Since its first description, the two isoforms of the receptor, membrane and soluble (mTREM-1 and sTREM-1, respectively) have been largely explored in the immunopathogenesis of several bacterial diseases and sepsis. The role of the receptor in these scenarios seems to be at least partly dependent on the source/type of bacteria, host and context. As uncontrolled inflammation is a result of several bacterial infections, the inhibition of the receptor has been considered as a promising approach to treat such conditions. Further, sTREM-1 has been explored as a biomarker for diagnosis and/or prognosis of several bacterial diseases. Therefore, this review aims to provide an updated insight into how the receptor influences and is influenced by bacterial infections, highlighting the advances regarding the use/manipulation of TREM-1 isoforms in biomedical research and clinical practice.
Collapse
Affiliation(s)
| | | | - Marcelle Silva-Sales
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | | |
Collapse
|
16
|
Vega MA, Simón-Fuentes M, González de la Aleja A, Nieto C, Colmenares M, Herrero C, Domínguez-Soto Á, Corbí ÁL. MAFB and MAF Transcription Factors as Macrophage Checkpoints for COVID-19 Severity. Front Immunol 2020; 11:603507. [PMID: 33312178 PMCID: PMC7708330 DOI: 10.3389/fimmu.2020.603507] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/19/2020] [Indexed: 01/10/2023] Open
Abstract
Defective IFN production and exacerbated inflammatory and pro-fibrotic responses are hallmarks of SARS-CoV-2 infection in severe COVID-19. Based on these hallmarks, and considering the pivotal role of macrophages in COVID-19 pathogenesis, we hypothesize that the transcription factors MAFB and MAF critically contribute to COVID-19 progression by shaping the response of macrophages to SARS-CoV-2. Our proposal stems from the recent identification of pathogenic lung macrophage subsets in severe COVID-19, and takes into consideration the previously reported ability of MAFB to dampen IFN type I production, as well as the critical role of MAFB and MAF in the acquisition and maintenance of the transcriptional signature of M-CSF-conditioned human macrophages. Solid evidences are presented that link overexpression of MAFB and silencing of MAF expression with clinical and biological features of severe COVID-19. As a whole, we propose that a high MAFB/MAF expression ratio in lung macrophages could serve as an accurate diagnostic tool for COVID-19 progression. Indeed, reversing the macrophage MAFB/MAF expression ratio might impair the exacerbated inflammatory and profibrotic responses, and restore the defective IFN type I production, thus becoming a potential strategy to limit severity of COVID-19.
Collapse
Affiliation(s)
- Miguel A. Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | | | | | | | | | | | | | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
17
|
Yasmeen F, Seo H, Javaid N, Kim MS, Choi S. Therapeutic Interventions into Innate Immune Diseases by Means of Aptamers. Pharmaceutics 2020; 12:pharmaceutics12100955. [PMID: 33050544 PMCID: PMC7600108 DOI: 10.3390/pharmaceutics12100955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/03/2020] [Accepted: 10/04/2020] [Indexed: 12/25/2022] Open
Abstract
The immune system plays a crucial role in the body's defense system against various pathogens, such as bacteria, viruses, and parasites, as well as recognizes non-self- and self-molecules. The innate immune system is composed of special receptors known as pattern recognition receptors, which play a crucial role in the identification of pathogen-associated molecular patterns from diverse microorganisms. Any disequilibrium in the activation of a particular pattern recognition receptor leads to various inflammatory, autoimmune, or immunodeficiency diseases. Aptamers are short single-stranded deoxyribonucleic acid or ribonucleic acid molecules, also termed "chemical antibodies," which have tremendous specificity and affinity for their target molecules. Their features, such as stability, low immunogenicity, ease of manufacturing, and facile screening against a target, make them preferable as therapeutics. Immune-system-targeting aptamers have a great potential as a targeted therapeutic strategy against immune diseases. This review summarizes components of the innate immune system, aptamer production, pharmacokinetic characteristics of aptamers, and aptamers related to innate-immune-system diseases.
Collapse
|
18
|
Samaddar S, Marnin L, Butler LR, Pedra JHF. Immunometabolism in Arthropod Vectors: Redefining Interspecies Relationships. Trends Parasitol 2020; 36:807-815. [PMID: 32819827 PMCID: PMC7897511 DOI: 10.1016/j.pt.2020.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/13/2020] [Accepted: 07/18/2020] [Indexed: 02/08/2023]
Abstract
Metabolism influences biochemical networks, and arthropod vectors are endowed with an immune system that affects microbial acquisition, persistence, and transmission to humans and other animals. Here, we aim to persuade the scientific community to expand their interests in immunometabolism beyond mammalian hosts and towards arthropod vectors. Immunometabolism investigates the interplay of metabolism and immunology. We provide a conceptual framework for investigators from diverse disciplines and indicate that relationships between microbes, mammalian hosts and their hematophagous arthropods may result in cost-effective (mutualism) or energetically expensive (parasitism) interactions. We argue that disparate resource allocations between species may partially explain why some microbes act as pathogens when infecting humans and behave as mutualistic or commensal organisms when colonizing arthropod vectors.
Collapse
Affiliation(s)
- Sourabh Samaddar
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
19
|
Bacterial virulence mediated by orthogonal post-translational modification. Nat Chem Biol 2020; 16:1043-1051. [PMID: 32943788 DOI: 10.1038/s41589-020-0638-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/30/2020] [Indexed: 12/28/2022]
Abstract
Many bacterial pathogens secrete virulence factors, also known as effector proteins, directly into host cells. These effectors suppress pro-inflammatory host signaling while promoting bacterial infection. A particularly interesting subset of effectors post-translationally modify host proteins using novel chemistry that is not otherwise found in the mammalian proteome, which we refer to as 'orthogonal post-translational modification' (oPTM). In this Review, we profile oPTM chemistry for effectors that catalyze serine/threonine acetylation, phosphate β-elimination, phosphoribosyl-linked ubiquitination, glutamine deamidation, phosphocholination, cysteine methylation, arginine N-acetylglucosaminylation, and glutamine ADP-ribosylation on host proteins. AMPylation, a PTM that could be considered orthogonal until only recently, is also discussed. We further highlight known cellular targets of oPTMs and their resulting biological consequences. Developing a complete understanding of oPTMs and the host cell processes they hijack will illuminate critical steps in the infection process, which can be harnessed for a variety of therapeutic, diagnostic, and synthetic applications.
Collapse
|
20
|
Kim SM, DeFazio JR, Hyoju SK, Sangani K, Keskey R, Krezalek MA, Khodarev NN, Sangwan N, Christley S, Harris KG, Malik A, Zaborin A, Bouziat R, Ranoa DR, Wiegerinck M, Ernest JD, Shakhsheer BA, Fleming ID, Weichselbaum RR, Antonopoulos DA, Gilbert JA, Barreiro LB, Zaborina O, Jabri B, Alverdy JC. Fecal microbiota transplant rescues mice from human pathogen mediated sepsis by restoring systemic immunity. Nat Commun 2020; 11:2354. [PMID: 32393794 PMCID: PMC7214422 DOI: 10.1038/s41467-020-15545-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
Death due to sepsis remains a persistent threat to critically ill patients confined to the intensive care unit and is characterized by colonization with multi-drug-resistant healthcare-associated pathogens. Here we report that sepsis in mice caused by a defined four-member pathogen community isolated from a patient with lethal sepsis is associated with the systemic suppression of key elements of the host transcriptome required for pathogen clearance and decreased butyrate expression. More specifically, these pathogens directly suppress interferon regulatory factor 3. Fecal microbiota transplant (FMT) reverses the course of otherwise lethal sepsis by enhancing pathogen clearance via the restoration of host immunity in an interferon regulatory factor 3-dependent manner. This protective effect is linked to the expansion of butyrate-producing Bacteroidetes. Taken together these results suggest that fecal microbiota transplantation may be a treatment option in sepsis associated with immunosuppression. Sepsis due to multidrug resistant pathogens is the most common cause of death in intensive care units. Here, the authors report that fecal microbiota transplant (FMT) can rescue mice from lethal sepsis of pathogens isolated from stool of a critically ill patient and show that FMT reverses the immunosuppressive effect induced by the pathogen community.
Collapse
Affiliation(s)
- Sangman M Kim
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA.,Department of Biology, University of San Francisco, San Francisco, CA, USA
| | - Jennifer R DeFazio
- Department of Surgery, University of Chicago, Chicago, IL, USA.,Department of Surgery, Columbia University, New York, NY, USA
| | - Sanjiv K Hyoju
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Kishan Sangani
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Robert Keskey
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Surgery, University of Chicago, Chicago, IL, USA
| | | | - Nikolai N Khodarev
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Naseer Sangwan
- Department of Surgery, University of Chicago, Chicago, IL, USA.,Argonne National Laboratory, Argonne, IL, USA
| | - Scott Christley
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | | | - Ankit Malik
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Romain Bouziat
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Diana R Ranoa
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Mara Wiegerinck
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Jordan D Ernest
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Irma D Fleming
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Dionysios A Antonopoulos
- Department of Medicine, University of Chicago, Chicago, IL, USA.,Argonne National Laboratory, Argonne, IL, USA
| | - Jack A Gilbert
- Department of Surgery, University of Chicago, Chicago, IL, USA.,Argonne National Laboratory, Argonne, IL, USA
| | - Luis B Barreiro
- Department of Medicine, University of Chicago, Chicago, IL, USA.,Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Olga Zaborina
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA. .,Department of Medicine, University of Chicago, Chicago, IL, USA. .,Department of Pathology, University of Chicago, Chicago, IL, USA. .,Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, IL, USA.
| | - John C Alverdy
- Department of Surgery, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
21
|
Cai L, Tong J, Zhang Z, Zhang Y, Jiang L, Hou X, Zhang H. Staphylococcus aureus-induced proteomic changes in the mammary tissue of rats: A TMT-based study. PLoS One 2020; 15:e0231168. [PMID: 32365127 PMCID: PMC7197811 DOI: 10.1371/journal.pone.0231168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/17/2020] [Indexed: 01/29/2023] Open
Abstract
Staphylococcus aureus is one of the most important pathogens causing mastitis in dairy cows. The objective of this study was to establish a rat model of mastitis induced by S. aureus infection and to explore changes in the proteomes of mammary tissue in different udder states, providing a better understanding of the host immune response to S. aureus mastitis. On day 3 post-partum, 6 rats were randomly divided into two groups (n = 3), with either 100 μL of PBS (blank group) or a S. aureus suspension containing 2×107 CFU·mL−1 (challenge group) infused into the mammary gland duct. After 24 h of infection, the rats were sacrificed, and mammary gland tissue was collected. Tandem mass tag (TMT)-based technology was applied to compare the proteomes of healthy and mastitic mammary tissues. Compared with the control group, the challenge group had 555 proteins with significant differences in expression, of which 428 were significantly upregulated (FC>1.2 and p<0.05) and 127 were downregulated (FC>0.83 and p<0.05 or p<0.01). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that upregulated differentially significant expressed proteins (DSEPs) were associated with mainly immune responses, including integrin alpha M, inter-α-trypsin inhibitor heavy chain 4, and alpha-2-macroglobulin. This study is the first in which a rat model of S. aureus-induced mastitis was used to explore the proteins related to mastitis in dairy cows by TMT technology, providing a model for replication of dairy cow S. aureus-induced mastitis experiments.
Collapse
Affiliation(s)
- Lirong Cai
- Beijing Key Laboratory of Dairy Cow Nutrition, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Jinjin Tong
- Beijing Key Laboratory of Dairy Cow Nutrition, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Zhaonan Zhang
- Beijing Key Laboratory of Dairy Cow Nutrition, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yonghong Zhang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Linshu Jiang
- Beijing Key Laboratory of Dairy Cow Nutrition, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Xiaolin Hou
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Hua Zhang
- Beijing Key Laboratory of Dairy Cow Nutrition, Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
- * E-mail:
| |
Collapse
|
22
|
Disruption of the cpsE and endA Genes Attenuates Streptococcus pneumoniae Virulence: Towards the Development of a Live Attenuated Vaccine Candidate. Vaccines (Basel) 2020; 8:vaccines8020187. [PMID: 32326482 PMCID: PMC7349068 DOI: 10.3390/vaccines8020187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
The majority of deaths due to Streptococcus pneumoniae infections are in developing countries. Although polysaccharide-based pneumococcal vaccines are available, newer types of vaccines are needed to increase vaccine affordability, particularly in developing countries, and to provide broader protection across all pneumococcal serotypes. To attenuate pneumococcal virulence with the aim of engineering candidate live attenuated vaccines (LAVs), we constructed knockouts in S. pneumoniae D39 of one of the capsular biosynthetic genes, cpsE that encodes glycosyltransferase, and the endonuclease gene, endA, that had been implicated in the uptake of DNA from the environment as well as bacterial escape from neutrophil-mediated killing. The cpsE gene knockout significantly lowered peak bacterial density, BALB/c mice nasopharyngeal (NP) colonisation but increased biofilm formation when compared to the wild-type D39 strain as well as the endA gene knockout mutant. All constructed mutant strains were able to induce significantly high serum and mucosal antibody response in BALB/c mice. However, the cpsE-endA double mutant strain, designated SPEC, was able to protect mice from high dose mucosal challenge of the D39 wild-type. Furthermore, SPEC showed 23-fold attenuation of virulence compared to the wild-type. Thus, the cpsE-endA double-mutant strain could be a promising candidate for further development of a LAV for S. pneumoniae.
Collapse
|
23
|
Degos C, Hysenaj L, Gonzalez‐Espinoza G, Arce‐Gorvel V, Gagnaire A, Papadopoulos A, Pasquevich KA, Méresse S, Cassataro J, Mémet S, Gorvel J. Omp25‐dependent engagement of SLAMF1 byBrucella abortusin dendritic cells limits acute inflammation and favours bacterial persistence in vivo. Cell Microbiol 2020; 22:e13164. [DOI: 10.1111/cmi.13164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Clara Degos
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Lisiena Hysenaj
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | | | - Vilma Arce‐Gorvel
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Aurélie Gagnaire
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Alexia Papadopoulos
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Karina Alejandra Pasquevich
- Instituto de Investigaciones BiotecnológicasUniversidad Nacional de San Martín (UNSAM)‐CONICET Buenos Aires Argentina
| | - Stéphane Méresse
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Juliana Cassataro
- Instituto de Investigaciones BiotecnológicasUniversidad Nacional de San Martín (UNSAM)‐CONICET Buenos Aires Argentina
| | - Sylvie Mémet
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| | - Jean‐Pierre Gorvel
- CNRS, INSERM, CIML, Centre d'Immunologie de Marseille‐LuminyAix‐Marseille University Marseille France
| |
Collapse
|
24
|
Ariana A, Alturki NA, Hajjar S, Stumpo DJ, Tiedje C, Alnemri ES, Gaestel M, Blackshear PJ, Sad S. Tristetraprolin regulates necroptosis during tonic Toll-like receptor 4 (TLR4) signaling in murine macrophages. J Biol Chem 2020; 295:4661-4672. [PMID: 32094226 DOI: 10.1074/jbc.ra119.011633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/10/2020] [Indexed: 11/06/2022] Open
Abstract
The necrosome is a protein complex required for signaling in cells that results in necroptosis, which is also dependent on tumor necrosis factor receptor (TNF-R) signaling. TNFα promotes necroptosis, and its expression is facilitated by mitogen-activated protein (MAP) kinase-activated protein kinase 2 (MK2) but is inhibited by the RNA-binding protein tristetraprolin (TTP, encoded by the Zfp36 gene). We have stimulated murine macrophages from WT, MyD88 -/-, Trif -/-, MyD88 -/- Trif -/-, MK2 -/-, and Zfp36 -/- mice with graded doses of lipopolysaccharide (LPS) and various inhibitors to evaluate the role of various genes in Toll-like receptor 4 (TLR4)-induced necroptosis. Necrosome signaling, cytokine production, and cell death were evaluated by immunoblotting, ELISA, and cell death assays, respectively. We observed that during TLR4 signaling, necrosome activation is mediated through the adaptor proteins MyD88 and TRIF, and this is inhibited by MK2. In the absence of MK2-mediated necrosome activation, lipopolysaccharide-induced TNFα expression was drastically reduced, but MK2-deficient cells became highly sensitive to necroptosis even at low TNFα levels. In contrast, during tonic TLR4 signaling, WT cells did not undergo necroptosis, even when MK2 was disabled. Of note, necroptosis occurred only in the absence of TTP and was mediated by the expression of TNFα and activation of JUN N-terminal kinase (JNK). These results reveal that TTP plays an important role in inhibiting TNFα/JNK-induced necrosome signaling and resultant cytotoxicity.
Collapse
Affiliation(s)
- Ardeshir Ariana
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Norah A Alturki
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Stephanie Hajjar
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Deborah J Stumpo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Christopher Tiedje
- Department of Cellular and Molecular Medicine, University of Copenhagen, The Maersk Tower, 7.3, Blegdamsvej 3B, Copenhagen DK-2200, Denmark.,Institute of Cell Biochemistry, Hannover Medical School, Germany, 30623
| | - Emad S Alnemri
- Thomas Jefferson University, Department of Biochemistry and Molecular Biology, Philadelphia, Pennsylvania 19107
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Germany, 30623
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Subash Sad
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada .,University of Ottawa, Ottawa Centre for Infection, Immunity and Inflammation, Ontario K1H 8M5, Canada
| |
Collapse
|
25
|
Fu X, Yang Y. WEDeepT3: predicting type III secreted effectors based on word embedding and deep learning. QUANTITATIVE BIOLOGY 2019. [DOI: 10.1007/s40484-019-0184-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Zhang J, Tao J, Ling Y, Li F, Zhu X, Xu L, Wang M, Zhang S, McCall CE, Liu TF. Switch of NAD Salvage to de novo Biosynthesis Sustains SIRT1-RelB-Dependent Inflammatory Tolerance. Front Immunol 2019; 10:2358. [PMID: 31681271 PMCID: PMC6797595 DOI: 10.3389/fimmu.2019.02358] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/19/2019] [Indexed: 12/14/2022] Open
Abstract
A typical inflammatory response sequentially progresses from pro-inflammatory, immune suppressive to inflammatory repairing phases. Although the physiological inflammatory response resolves in time, severe acute inflammation usually sustains immune tolerance and leads to high mortality, yet the underlying mechanism is not completely understood. Here, using the leukemia-derived THP-1 human monocytes, healthy and septic human peripheral blood mononuclear cells (PBMC), we report that endotoxin dose-dependent switch of nicotinamide adenine dinucleotide (NAD) biosynthesis pathways sustain immune tolerant status. Low dose endotoxin triggered nicotinamide phosphoribosyltransferase (NAMPT)-dependent NAD salvage activity to adapt pro-inflammation. In contrast, high dose endotoxin drove a shift of NAD synthesis pathway from early NAMPT-dependent NAD salvage to late indoleamine 2,3-dioxygenase-1 (IDO1)-dependent NAD de novo biosynthesis, leading to persistent immune suppression. This is resulted from the IDO1-dependent expansion of nuclear NAD pool and nuclear NAD-dependent prolongation of sirtuin1 (SIRT1)-directed epigenetics of immune tolerance. Inhibition of IDO1 activity predominantly decreased nuclear NAD level, which promoted sequential dissociations of immunosuppressive SIRT1 and RelB from the promoter of pro-inflammatory TNF-α gene and broke endotoxin tolerance. Thus, NAMPT-NAD-SIRT1 axis adapts pro-inflammation, but IDO1-NAD-SIRT1-RelB axis sustains endotoxin tolerance during acute inflammatory response. Remarkably, in contrast to the prevention of sepsis death of animal model by IDO1 inhibition before sepsis initiation, we demonstrated that the combination therapy of IDO1 inhibition by 1-methyl-D-tryptophan (1-MT) and tryptophan supplementation rather than 1-MT administration alone after sepsis onset rescued sepsis animals, highlighting the translational significance of tryptophan restoration in IDO1 targeting therapy of severe inflammatory diseases like sepsis.
Collapse
Affiliation(s)
- Jingpu Zhang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jie Tao
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yun Ling
- Department of Infection Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Feng Li
- Department of Critical Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xuewei Zhu
- Molecular Medicine Section, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Li Xu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Mei Wang
- Department of Critical Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shuye Zhang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Charles E. McCall
- Molecular Medicine Section, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Tie Fu Liu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Molecular Medicine Section, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
27
|
Carter RA, Pan K, Harville EW, McRitchie S, Sumner S. Metabolomics to reveal biomarkers and pathways of preterm birth: a systematic review and epidemiologic perspective. Metabolomics 2019; 15:124. [PMID: 31506796 PMCID: PMC7805080 DOI: 10.1007/s11306-019-1587-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Most known risk factors for preterm birth, a leading cause of infant morbidity and mortality, are not modifiable. Advanced molecular techniques are increasingly being applied to identify biomarkers and pathways important in disease development and progression. OBJECTIVES We review the state of the literature and assess it from an epidemiologic perspective. METHODS PubMed, Embase, CINAHL, and Cochrane Central were searched on January 31, 2019 for original articles published after 1998 that utilized an untargeted metabolomic approach to identify markers of preterm birth. Eligible manuscripts were peer-reviewed and included original data from untargeted metabolomics analyses of maternal tissue derived from human studies designed to determine mechanisms and predictors of preterm birth. RESULTS Of 2823 results, 14 articles met the inclusion requirements. There was little consistency in study design, outcome definition, type of biospecimen, or the inclusion of covariates and confounding factors, and few consistent associations with metabolites were identified in this review. CONCLUSION Studies to date on metabolomic predictors of preterm birth are highly heterogeneous in both methodology and resulting metabolite identification. There is an urgent need for larger studies in well-defined populations, to determine biomarkers predictive of preterm birth, and to reveal mechanisms and targets for development of intervention strategies.
Collapse
Affiliation(s)
- R A Carter
- Department of Epidemiology, Tulane School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA, 70112, USA
| | - K Pan
- Department of Epidemiology, Tulane School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA, 70112, USA.
| | - E W Harville
- Department of Epidemiology, Tulane School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA, 70112, USA
| | - S McRitchie
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, USA
| | - S Sumner
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, USA
| |
Collapse
|
28
|
Nowill AE, Fornazin MC, Spago MC, Dorgan Neto V, Pinheiro VRP, Alexandre SSS, Moraes EO, Souza GHMF, Eberlin MN, Marques LA, Meurer EC, Franchi GC, de Campos-Lima PO. Immune Response Resetting in Ongoing Sepsis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:1298-1312. [PMID: 31358659 PMCID: PMC6697741 DOI: 10.4049/jimmunol.1900104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/28/2019] [Indexed: 01/03/2023]
Abstract
Cure of severe infections, sepsis, and septic shock with antimicrobial drugs is a challenge because morbidity and mortality in these conditions are essentially caused by improper immune response. We have tested the hypothesis that repeated reactivation of established memory to pathogens may reset unfavorable immune responses. We have chosen for this purpose a highly stringent mouse model of polymicrobial sepsis by cecum ligation and puncture. Five weeks after priming with a diverse Ag pool, high-grade sepsis was induced in C57BL/6j mice that was lethal in 24 h if left untreated. Antimicrobial drug (imipenem) alone rescued 9.7% of the animals from death, but >5-fold higher cure rate could be achieved by combining imipenem and two rechallenges with the Ag pool (p < 0.0001). Antigenic stimulation fine-tuned the immune response in sepsis by contracting the total CD3+ T cell compartment in the spleen and disengaging the hyperactivation state in the memory T subsets, most notably CD8+ T cells, while preserving the recovery of naive subsets. Quantitative proteomics/lipidomics analyses revealed that the combined treatment reverted the molecular signature of sepsis for cytokine storm, and deregulated inflammatory reaction and proapoptotic environment, as well as the lysophosphatidylcholine/phosphatidylcholine ratio. Our results showed the feasibility of resetting uncontrolled hyperinflammatory reactions into ordered hypoinflammatory responses by memory reactivation, thereby reducing morbidity and mortality in antibiotic-treated sepsis. This beneficial effect was not dependent on the generation of a pathogen-driven immune response itself but rather on the reactivation of memory to a diverse Ag pool that modulates the ongoing response.
Collapse
Affiliation(s)
- Alexandre E Nowill
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas 13083-888, Brazil;
| | - Márcia C Fornazin
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas 13083-888, Brazil
| | - Maria C Spago
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas 13083-888, Brazil
| | - Vicente Dorgan Neto
- Surgery Department, Santa Casa School of Medical Sciences, São Paulo 01221-020, Brazil
| | - Vitória R P Pinheiro
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas 13083-888, Brazil
| | - Simônia S S Alexandre
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas 13083-888, Brazil;
| | - Edgar O Moraes
- School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, Brazil
| | - Gustavo H M F Souza
- Mass Spectrometry Research and Development Laboratory, Health Sciences Department, Waters Corporation, Barueri 06455-020, Brazil
| | - Marcos N Eberlin
- School of Engineering, Mackenzie Presbyterian University, São Paulo 01302-907, Brazil
| | - Lygia A Marques
- Thomson Mass Spectrometry Laboratory, Institute of Chemistry, State University of Campinas, Campinas 13083-859, Brazil; and
| | - Eduardo C Meurer
- Thomson Mass Spectrometry Laboratory, Institute of Chemistry, State University of Campinas, Campinas 13083-859, Brazil; and
| | - Gilberto C Franchi
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas 13083-888, Brazil
| | | |
Collapse
|
29
|
Ceciliani F, Lecchi C. The Immune Functions of α 1 Acid Glycoprotein. Curr Protein Pept Sci 2019; 20:505-524. [PMID: 30950347 DOI: 10.2174/1389203720666190405101138] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
α1-acid glycoprotein (orosomucoid, AGP) is an Acute Phase Protein produced by liver and peripheral tissues in response to systemic reaction to inflammation. AGP functions have been studied mostly in human, cattle and fish, although the protein has been also found in many mammalian species and birds. AGP fulfils at least two set of functions, which are apparently different from each other but in fact intimately linked. On one hand, AGP is an immunomodulatory protein. On the other hand, AGP is one of the most important binding proteins in plasma and, beside modulating pharmacokinetics and pharmacodynamics of many drugs, it is also able to bind and transport several endogen ligands related to inflammation. The focus of this review is the immunomodulatory activity of AGP. This protein regulates every single event related to inflammation, including binding of pathogens and modulating white blood cells activity throughout the entire leukocyte attacking sequence. The regulation of AGP activity is complex: the inflammation induces not only an increase in AGP serum concentration, but also a qualitative change in its carbohydrate moiety, generating a multitude of glycoforms, each of them with different, and sometimes opposite and contradictory, activities. We also present the most recent findings about the relationship between AGP and adipose tissue: AGP interacts with leptin receptor and, given its immunomodulatory function, it may be included among the potential players in the field of immunometabolism.
Collapse
Affiliation(s)
- Fabrizio Ceciliani
- Department of Veterinary Medicine, Universita degli Studi di Milano, Milano, Italy
| | - Cristina Lecchi
- Department of Veterinary Medicine, Universita degli Studi di Milano, Milano, Italy
| |
Collapse
|
30
|
David SC, Laan Z, Minhas V, Chen AY, Davies J, Hirst TR, McColl SR, Alsharifi M, Paton JC. Enhanced safety and immunogenicity of a pneumococcal surface antigen A mutant whole-cell inactivated pneumococcal vaccine. Immunol Cell Biol 2019; 97:726-739. [PMID: 31050022 DOI: 10.1111/imcb.12257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/21/2019] [Accepted: 04/29/2019] [Indexed: 01/14/2023]
Abstract
Existing capsular polysaccharide-based vaccines against pneumococcal disease are highly effective against vaccine-included serotypes, but they are unable to combat serotype replacement. We have developed a novel pneumococcal vaccine that confers serotype-independent protection, and could therefore constitute a "universal" vaccine formulation. This preparation is comprised of whole un-encapsulated pneumococci inactivated with gamma irradiation (γ-PN), and we have previously reported induction of cross-reactive immunity after nonadjuvanted intranasal vaccination. To further enhance vaccine immunogenicity and safety, we modified the pneumococcal vaccine strain to induce a stressed state during growth. Specifically, the substrate binding component of the psaBCA operon for manganese import was mutated to create a pneumococcal surface antigen A (psaA) defective vaccine strain. psaA mutation severely attenuated the growth of the vaccine strain in vitro without negatively affecting pneumococcal morphology, thereby enhancing vaccine safety. In addition, antibodies raised against vaccine preparations based on the modified strain [γ-PN(ΔPsaA)] showed more diversified reactivity to wild-type pneumococcal challenge strains compared to those induced by the original formulation. The modified vaccine also induced comparable protective TH 17 responses in the lung, and conferred greater protection against lethal heterologous pneumococcal challenge. Overall, the current study demonstrates successful refinement of a serotype-independent pneumococcal vaccine candidate to enhance safety and immunogenicity.
Collapse
Affiliation(s)
- Shannon C David
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Zoe Laan
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Vikrant Minhas
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Austen Y Chen
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Justin Davies
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Timothy R Hirst
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.,GPN Vaccines Pty Ltd, Yarralumla, ACT, Australia.,Gamma Vaccines Pty Ltd, Yarralumla, ACT, Australia
| | - Shaun R McColl
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Mohammed Alsharifi
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.,GPN Vaccines Pty Ltd, Yarralumla, ACT, Australia.,Gamma Vaccines Pty Ltd, Yarralumla, ACT, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, and Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.,GPN Vaccines Pty Ltd, Yarralumla, ACT, Australia
| |
Collapse
|
31
|
Zhang H, Jiang H, Fan Y, Chen Z, Li M, Mao Y, Karrow NA, Loor JJ, Moore S, Yang Z. Transcriptomics and iTRAQ-Proteomics Analyses of Bovine Mammary Tissue with Streptococcus agalactiae-Induced Mastitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:11188-11196. [PMID: 30096236 DOI: 10.1021/acs.jafc.8b02386] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Mastitis is a highly prevalent disease in dairy cows that causes large economic losses. Streptococcus agalactiae is a common contagious pathogen and a major cause of bovine mastitis. The immune response to intramammary infection with S. agalactiae in dairy cows is a very complex biological process. To understand the host immune response to S. agalactiae-induced mastitis, mammary gland of lactating Chinese Holstein cows was challenged with S. agalactiae via nipple tube perfusion. Visual inspection, analysis of milk somatic cell counts, histopathology, and transmission electron microscopy of mammary tissue were performed to confirm S. agalactiae-induced mastitis. Microarray and isobaric tags for relative and absolute quantitation (iTRAQ) were used to compare the transcriptomes and proteomes of healthy and mastitic mammary tissue. Compared with healthy tissue, a total of 129 differentially expressed genes (DEGs, fold change >2, p < 0.05) and 144 differentially expressed proteins (DEPs, fold change >1.2, p < 0.05) were identified in mammary tissue from S. agalactiae-challenged cows. Among the concordant 18 DEGs/DEPs, immunoglobulin M precursor, cathelicidin-7 precursor, integrin alpha-5, and complement C4-A-like isoform X1 were associated with mastitis. Intramammary infection with S. agalactiae triggered a complex host innate immune response that involved complement and coagulation cascades, ECM-receptor interaction, focal adhesion, and phagosome and bacterial invasion of epithelial cells pathways. These results provide candidate genes or proteins for further studies in the context of prevention and targeted treatment of bovine mastitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Niel A Karrow
- Department of Animal Biosciences , University of Guelph , Guelph N1G 2W1 , Canada
| | - Juan J Loor
- Department of Animal Sciences & Division of Nutritional Sciences , University of Illinois , Urbana , Illinois 61801 , United States
| | - Stephen Moore
- Centre for Animal Science , University of Queensland , Saint Luci , Queensland 4072a , Australia
| | | |
Collapse
|
32
|
Vadstein O, Attramadal KJK, Bakke I, Forberg T, Olsen Y, Verdegem M, Giatsis C, Skjermo J, Aasen IM, Gatesoupe FJ, Dierckens K, Sorgeloos P, Bossier P. Managing the Microbial Community of Marine Fish Larvae: A Holistic Perspective for Larviculture. Front Microbiol 2018; 9:1820. [PMID: 30210457 PMCID: PMC6119882 DOI: 10.3389/fmicb.2018.01820] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022] Open
Abstract
The availability of high-quality juveniles is a bottleneck in the farming of many marine fish species. Detrimental larvae-microbe interactions are a main reason for poor viability and quality in larval rearing. In this review, we explore the microbial community of fish larvae from an ecological and eco-physiological perspective, with the aim to develop the knowledge basis for microbial management. The larvae are exposed to a huge number of microbes from external and internal sources in intensive aquaculture, but their relative importance depend on the rearing technology used (especially flow-through vs. recirculating systems) and the retention time of the water in the fish tanks. Generally, focus has been on microbes entering the system, but microbes from growth within the system is normally a substantial part of the microbes encountered by larvae. Culture independent methods have revealed an unexpected high richness of bacterial species associated with larvae, with 100-250 operational taxonomic units associated with one individual. The microbiota of larvae changes rapidly until metamorphosis, most likely due to changes in the selection pressure in the digestive tract caused by changes in host-microbe and microbe-microbe interactions. Even though the microbiota of larvae is distinctly different from the microbiota of the water and the live food, the microbiota of the water strongly affects the microbiota of the larvae. We are in the early phase of understanding larvae-microbe interactions in vivo, but some studies with other animals than fish emphasize that we so far have underestimated the complexity of these interactions. We present examples demonstrating the diversity of these interactions. A large variety of microbial management methods exist, focusing on non-selective reduction of microbes, selective enhancement of microbes, and on improvement of the resistance of larvae against microbes. However, relatively few methods have been studied extensively. We believe that there is a lot to gain by increasing the diversity of approaches for microbial management. As many microbial management methods are perturbations of the microbial community, we argue that ecological theory is needed to foresee and test for longer term consequences in microbe-microbe and microbe-larvae interactions. We finally make some recommendations for future research and development.
Collapse
Affiliation(s)
- Olav Vadstein
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kari J. K. Attramadal
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingrid Bakke
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Torunn Forberg
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yngvar Olsen
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Marc Verdegem
- Aquaculture and Fisheries Group, Wageningen University, Wageningen, Netherlands
| | - Cristos Giatsis
- Aquaculture and Fisheries Group, Wageningen University, Wageningen, Netherlands
| | - Jorunn Skjermo
- Department of Environment and New Resources, SINTEF Ocean, Trondheim, Norway
| | - Inga M. Aasen
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | | | - Kristof Dierckens
- Faculty of Bioscience Engineering, Laboratory of Aquaculture and Artemia Reference Center, Ghent University, Ghent, Belgium
| | - Patrick Sorgeloos
- Faculty of Bioscience Engineering, Laboratory of Aquaculture and Artemia Reference Center, Ghent University, Ghent, Belgium
| | - Peter Bossier
- Faculty of Bioscience Engineering, Laboratory of Aquaculture and Artemia Reference Center, Ghent University, Ghent, Belgium
| |
Collapse
|
33
|
Laguri C, Silipo A, Martorana AM, Schanda P, Marchetti R, Polissi A, Molinaro A, Simorre JP. Solid State NMR Studies of Intact Lipopolysaccharide Endotoxin. ACS Chem Biol 2018; 13:2106-2113. [PMID: 29965728 DOI: 10.1021/acschembio.8b00271] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lipopolysaccharides (LPS) are complex glycolipids forming the outside layer of Gram-negative bacteria. Their hydrophobic and heterogeneous nature greatly hampers their structural study in an environment similar to the bacterial surface. We have studied LPS purified from E. coli and pathogenic P. aeruginosa with long O-antigen polysaccharides assembled in solution as vesicles or elongated micelles. Solid-state NMR with magic-angle spinning permitted the identification of NMR signals arising from regions with different flexibilities in the LPS, from the lipid components to the O-antigen polysaccharides. Atomic scale data on the LPS enabled the study of the interaction of gentamicin antibiotic bound to P. aeruginosa LPS, for which we could confirm that a specific oligosaccharide is involved in the antibiotic binding. The possibility to study LPS alone and bound to a ligand when it is assembled in membrane-like structures opens great prospects for the investigation of proteins and antibiotics that specifically target such an important molecule at the surface of Gram-negative bacteria.
Collapse
Affiliation(s)
- Cedric Laguri
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | - Alba Silipo
- University of Naples Federico II, Department of Chemical Sciences, via cintia 4, Napoli, Italy
| | - Alessandra M. Martorana
- University of Milano, Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, Milano, Italy
| | - Paul Schanda
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | - Roberta Marchetti
- University of Naples Federico II, Department of Chemical Sciences, via cintia 4, Napoli, Italy
| | - Alessandra Polissi
- University of Milano, Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, Milano, Italy
| | - Antonio Molinaro
- University of Naples Federico II, Department of Chemical Sciences, via cintia 4, Napoli, Italy
| | | |
Collapse
|
34
|
Chambers KA, Abularrage NS, Scheck RA. Selectivity within a Family of Bacterial Phosphothreonine Lyases. Biochemistry 2018; 57:3790-3796. [PMID: 29792689 DOI: 10.1021/acs.biochem.8b00534] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Phosphothreonine lyases are bacterial effector proteins secreted into host cells to facilitate the infection process. This enzyme family catalyzes an irreversible elimination reaction that converts phosphothreonine or phosphoserine to dehydrobutyrine or dehydroalanine, respectively. Herein, we report a study of substrate selectivity for each of the four known phosphothreonine lyases. This was accomplished using a combination of mass spectrometry and enzyme kinetics assays for a series of phosphorylated peptides derived from the mitogen-activated protein kinase (MAPK) activation loop. These studies provide the first experimental evidence that VirA, a putative phosphothreonine lyase identified through homology, is indeed capable of catalyzing phosphate elimination. These studies further demonstrate that OspF is the most promiscuous phosphothreonine lyase, whereas SpvC is the most specific for the MAPK activation loop. Our studies reveal that phospholyases are dramatically more efficient at catalyzing elimination from phosphothreonine than from phosphoserine. Together, our data suggest that each enzyme likely has preferred substrates, either within the MAPK family or beyond. Fully understanding the extent of selectivity is key to understanding the impact of phosphothreonine lyases during bacterial infection and to exploiting their unique chemistry for a range of applications.
Collapse
Affiliation(s)
- Kaitlin A Chambers
- Department of Chemistry , Tufts University , 62 Talbot Avenue , Medford , Massachusetts 02155 , United States
| | - Nile S Abularrage
- Department of Chemistry , Tufts University , 62 Talbot Avenue , Medford , Massachusetts 02155 , United States
| | - Rebecca A Scheck
- Department of Chemistry , Tufts University , 62 Talbot Avenue , Medford , Massachusetts 02155 , United States
| |
Collapse
|
35
|
Tao J, Zhang J, Ling Y, McCall CE, Liu TF. Mitochondrial Sirtuin 4 Resolves Immune Tolerance in Monocytes by Rebalancing Glycolysis and Glucose Oxidation Homeostasis. Front Immunol 2018; 9:419. [PMID: 29593712 PMCID: PMC5854658 DOI: 10.3389/fimmu.2018.00419] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/15/2018] [Indexed: 12/30/2022] Open
Abstract
The goal of this investigation was to define the molecular mechanism underlying physiologic conversion of immune tolerance to resolution of the acute inflammatory response, which is unknown. An example of this knowledge gap and its clinical importance is the broad-based energy deficit and immunometabolic paralysis in blood monocytes from non-survivors of human and mouse sepsis that precludes sepsis resolution. This immunometabolic dysregulation is biomarked by ex vivo endotoxin tolerance to increased glycolysis and TNF-α expression. To investigate how tolerance switches to resolution, we adapted our previously documented models associated with acute inflammatory, immune, and metabolic reprogramming that induces endotoxin tolerance as a model of sepsis in human monocytes. We report here that mitochondrial sirtuin 4 (SIRT4) physiologically breaks tolerance and resolves acute inflammation in human monocytes by coordinately reprogramming of metabolism and bioenergetics. We find that increased SIRT4 mRNA and protein expression during immune tolerance counters the increase in pyruvate dehydrogenase kinase 1 (PDK1) and SIRT1 that promote tolerance by switching glucose-dependent support of immune resistance to fatty acid oxidation support of immune tolerance. By decreasing PDK1, pyruvate dehydrogenase complex reactivation rebalances mitochondrial respiration, and by decreasing SIRT1, SIRT4 represses fatty acid oxidation. The precise mechanism for the mitochondrial SIRT4 nuclear feedback is unclear. Our findings are consistent with a new concept in which mitochondrial SIRT4 directs the axis that controls anabolic and catabolic energy sources.
Collapse
Affiliation(s)
- Jie Tao
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jingpu Zhang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yun Ling
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Charles E McCall
- Molecular Medicine Section, Department of Internal Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Tie Fu Liu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.,Molecular Medicine Section, Department of Internal Medicine, Wake Forest University, Winston-Salem, NC, United States
| |
Collapse
|
36
|
Brenhouse HC, Danese A, Grassi-Oliveira R. Neuroimmune Impacts of Early-Life Stress on Development and Psychopathology. Curr Top Behav Neurosci 2018; 43:423-447. [PMID: 30003509 DOI: 10.1007/7854_2018_53] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Maltreatment and trauma in childhood, termed early-life stress (ELS), has long-term effects on the immune system. ELS impacts immune signaling at the time of exposure but also disrupts the developmental trajectory of certain immunological processes, both in the periphery and in the brain. One consequence of these early alterations is a heightened immune response to stressors later in life. However, chronic and sustained inflammatory response can also lead to excitotoxicity and prevent typical brain development. In this chapter, we discuss current progress toward understanding the contribution of neuroimmune signaling to ELS-attributable dysfunction or maladaptation with a focus on postnatal experiences. To do so we first present an operational definition of ELS. Then, we offer a brief overview of the immune system and neuroimmune development, followed by a section discussing the interaction between immunity, childhood trauma, and mental disorders in humans. We present evidence from animal models about immune alterations after ELS and discuss the ways in which ELS-induced immune changes ultimately affect brain and behavior, as well as the importance of individual differences and future directions in this field. Taken together, we submit that when encountered with ELS, some core brain circuits could develop differently via various mechanisms involving dysfunctional immune reprograming. However, given the remarkable plasticity of both the brain and the immune system, many of the deleterious effects of ELS may be mitigated with interventions that account for sex and target neuroimmune interactions over the lifespan.
Collapse
Affiliation(s)
| | - Andrea Danese
- Social, Genetic, and Developmental Psychiatry Centre, Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL), Graduate Program in Psychology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
37
|
Weinert LA, Welch JJ. Why Might Bacterial Pathogens Have Small Genomes? Trends Ecol Evol 2017; 32:936-947. [PMID: 29054300 DOI: 10.1016/j.tree.2017.09.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/31/2022]
Abstract
Bacteria that cause serious disease often have smaller genomes, and fewer genes, than their nonpathogenic, or less pathogenic relatives. Here, we review evidence for the generality of this association, and summarise the various reasons why the association might hold. We focus on the population genetic processes that might lead to reductive genome evolution, and show how several of these could be connected to pathogenicity. We find some evidence for most of the processes having acted in bacterial pathogens, including several different modes of genome reduction acting in the same lineage. We argue that predictable processes of genome evolution might not reflect any common underlying process.
Collapse
Affiliation(s)
- Lucy A Weinert
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK.
| | - John J Welch
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| |
Collapse
|
38
|
Abstract
Hundreds of different species colonize multicellular organisms making them "metaorganisms". A growing body of data supports the role of microbiota in health and in disease. Grasping the principles of host-microbiota interactions (HMIs) at the molecular level is important since it may provide insights into the mechanisms of infections. The crosstalk between the host and the microbiota may help resolve puzzling questions such as how a microorganism can contribute to both health and disease. Integrated superorganism networks that consider host and microbiota as a whole-may uncover their code, clarifying perhaps the most fundamental question: how they modulate immune surveillance. Within this framework, structural HMI networks can uniquely identify potential microbial effectors that target distinct host nodes or interfere with endogenous host interactions, as well as how mutations on either host or microbial proteins affect the interaction. Furthermore, structural HMIs can help identify master host cell regulator nodes and modules whose tweaking by the microbes promote aberrant activity. Collectively, these data can delineate pathogenic mechanisms and thereby help maximize beneficial therapeutics. To date, challenges in experimental techniques limit large-scale characterization of HMIs. Here we highlight an area in its infancy which we believe will increasingly engage the computational community: predicting interactions across kingdoms, and mapping these on the host cellular networks to figure out how commensal and pathogenic microbiota modulate the host signaling and broadly cross-species consequences.
Collapse
Affiliation(s)
- Emine Guven-Maiorov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Chung-Jung Tsai
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
- Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
Peterson LW, Philip NH, DeLaney A, Wynosky-Dolfi MA, Asklof K, Gray F, Choa R, Bjanes E, Buza EL, Hu B, Dillon CP, Green DR, Berger SB, Gough PJ, Bertin J, Brodsky IE. RIPK1-dependent apoptosis bypasses pathogen blockade of innate signaling to promote immune defense. J Exp Med 2017; 214:3171-3182. [PMID: 28855241 PMCID: PMC5679171 DOI: 10.1084/jem.20170347] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/19/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
RIPK1 regulates cytokine signaling and cell death during infection and inflammation. Peterson et al. show that RIPK1 kinase activity triggers apoptosis in response to bacterial pathogen blockade of innate immune signaling and that this pathway of effector-triggered immunity is critical for a successful antibacterial response. Many pathogens deliver virulence factors or effectors into host cells in order to evade host defenses and establish infection. Although such effector proteins disrupt critical cellular signaling pathways, they also trigger specific antipathogen responses, a process termed “effector-triggered immunity.” The Gram-negative bacterial pathogen Yersinia inactivates critical proteins of the NF-κB and MAPK signaling cascade, thereby blocking inflammatory cytokine production but also inducing apoptosis. Yersinia-induced apoptosis requires the kinase activity of receptor-interacting protein kinase 1 (RIPK1), a key regulator of cell death, NF-κB, and MAPK signaling. Through the targeted disruption of RIPK1 kinase activity, which selectively disrupts RIPK1-dependent cell death, we now reveal that Yersinia-induced apoptosis is critical for host survival, containment of bacteria in granulomas, and control of bacterial burdens in vivo. We demonstrate that this apoptotic response provides a cell-extrinsic signal that promotes optimal innate immune cytokine production and antibacterial defense, demonstrating a novel role for RIPK1 kinase–induced apoptosis in mediating effector-triggered immunity to circumvent pathogen inhibition of immune signaling.
Collapse
Affiliation(s)
- Lance W Peterson
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA.,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Naomi H Philip
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA.,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Alexandra DeLaney
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA.,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Meghan A Wynosky-Dolfi
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA.,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Kendra Asklof
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Falon Gray
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Ruth Choa
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA.,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Elisabet Bjanes
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA.,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Elisabeth L Buza
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Baofeng Hu
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | | | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | - Scott B Berger
- Host Defense Discovery Performance Unit, Infectious Disease Therapy Area Unit, GlaxoSmithKline, Collegeville, PA
| | - Peter J Gough
- Host Defense Discovery Performance Unit, Infectious Disease Therapy Area Unit, GlaxoSmithKline, Collegeville, PA
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA .,Institue for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
40
|
Korla K, Chandra N. A Systems Perspective of Signalling Networks in Host–Pathogen Interactions. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
41
|
Garib FY, Rizopulu AP, Kuchmiy AA, Garib VF. Inactivation of Inflammasomes by Pathogens Regulates Inflammation. BIOCHEMISTRY (MOSCOW) 2017; 81:1326-1339. [PMID: 27914458 DOI: 10.1134/s0006297916110109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammatory response is initiated and sustained by the action of quintessential pro-inflammatory cytokines of immune system namely IL-1β and IL-18. The maturation process of those cytokines is ensured by caspase-1 enzymatic activity, that is in turn is tightly controlled by multiprotein complexes called inflammasomes. Inflammasomes are activated in cells of innate immune system in response to recognition of conservative parts of microbes (pathogen-associated molecular patterns) or by sensing molecular signs of tissue damage (damage-associated molecular patterns). Inflammasome activation apart of cytokines secretion leads to pro-inflammatory cell death, so-called pyroptosis. That culminates in release of cytoplasmatic content of cells including cytokines and alarmins that boost immune response against pathogens, as well as pyroptosis destroys replicative niches of intracellular pathogens. During co-evolution with the host, bacterial and viral pathogens developed a range of molecular inhibitors targeting each step of inflammasome activation. In current review, we will discuss the latest knowledge of inflammasomes' signaling pathways and tricks that pathogens use to avoid immune recognition and clearance. Our better understanding of inflammasome inhibition by pathogens can lead to better therapeutic approaches for the treatment of infectious diseases.
Collapse
Affiliation(s)
- F Yu Garib
- Lomonosov Moscow State University, Biological Faculty, Moscow, 119991, Russia
| | | | | | | |
Collapse
|
42
|
Liu J, Xiang J, Li X, Blankson S, Zhao S, Cai J, Jiang Y, Redmond HP, Wang JH. NF-κB activation is critical for bacterial lipoprotein tolerance-enhanced bactericidal activity in macrophages during microbial infection. Sci Rep 2017; 7:40418. [PMID: 28079153 PMCID: PMC5227741 DOI: 10.1038/srep40418] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Tolerance to bacterial components represents an essential regulatory mechanism during bacterial infection. Bacterial lipoprotein (BLP)-induced tolerance confers protection against microbial sepsis by attenuating inflammatory responses and augmenting antimicrobial activity in innate phagocytes. It has been well-documented that BLP tolerance-attenuated proinflammatory cytokine production is associated with suppressed TLR2 signalling pathway; however, the underlying mechanism(s) involved in BLP tolerance-enhanced antimicrobial activity is unclear. Here we report that BLP-tolerised macrophages exhibited accelerated phagosome maturation and enhanced bactericidal activity upon bacterial infection, with upregulated expression of membrane-trafficking regulators and lysosomal enzymes. Notably, bacterial challenge resulted in a strong activation of NF-κB pathway in BLP-tolerised macrophages. Importantly, activation of NF-κB pathway is critical for BLP tolerance-enhanced antimicrobial activity, as deactivation of NF-κB in BLP-tolerised macrophages impaired phagosome maturation and intracellular killing of the ingested bacteria. Finally, activation of NF-κB pathway in BLP-tolerised macrophages was dependent on NOD1 and NOD2 signalling, as knocking-down NOD1 and NOD2 substantially inhibited bacteria-induced activation of NF-κB and overexpression of Rab10 and Acp5, two membrane-trafficking regulators and lysosomal enzymes contributed to BLP tolerance-enhanced bactericidal activity. These results indicate that activation of NF-κB pathway is essential for BLP tolerance-augmented antimicrobial activity in innate phagocytes and depends primarily on both NOD1 and NOD2.
Collapse
Affiliation(s)
- Jinghua Liu
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - Jing Xiang
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - Xue Li
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - Siobhan Blankson
- Department of Academic Surgery, University College Cork/National University of Ireland, Cork University Hospital, Cork, Ireland
| | - Shuqi Zhao
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - Junwei Cai
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - Yong Jiang
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - H Paul Redmond
- Department of Academic Surgery, University College Cork/National University of Ireland, Cork University Hospital, Cork, Ireland
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork/National University of Ireland, Cork University Hospital, Cork, Ireland
| |
Collapse
|
43
|
Jinawath N, Bunbanjerdsuk S, Chayanupatkul M, Ngamphaiboon N, Asavapanumas N, Svasti J, Charoensawan V. Bridging the gap between clinicians and systems biologists: from network biology to translational biomedical research. J Transl Med 2016; 14:324. [PMID: 27876057 PMCID: PMC5120462 DOI: 10.1186/s12967-016-1078-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/08/2016] [Indexed: 01/22/2023] Open
Abstract
With the wealth of data accumulated from completely sequenced genomes and other high-throughput experiments, global studies of biological systems, by simultaneously investigating multiple biological entities (e.g. genes, transcripts, proteins), has become a routine. Network representation is frequently used to capture the presence of these molecules as well as their relationship. Network biology has been widely used in molecular biology and genetics, where several network properties have been shown to be functionally important. Here, we discuss how such methodology can be useful to translational biomedical research, where scientists traditionally focus on one or a small set of genes, diseases, and drug candidates at any one time. We first give an overview of network representation frequently used in biology: what nodes and edges represent, and review its application in preclinical research to date. Using cancer as an example, we review how network biology can facilitate system-wide approaches to identify targeted small molecule inhibitors. These types of inhibitors have the potential to be more specific, resulting in high efficacy treatments with less side effects, compared to the conventional treatments such as chemotherapy. Global analysis may provide better insight into the overall picture of human diseases, as well as identify previously overlooked problems, leading to rapid advances in medicine. From the clinicians’ point of view, it is necessary to bridge the gap between theoretical network biology and practical biomedical research, in order to improve the diagnosis, prevention, and treatment of the world’s major diseases.
Collapse
Affiliation(s)
- Natini Jinawath
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.,Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sacarin Bunbanjerdsuk
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Maneerat Chayanupatkul
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nuttapong Ngamphaiboon
- Medical Oncology Unit, Department of Medicine Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nithi Asavapanumas
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Jisnuson Svasti
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.,Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| | - Varodom Charoensawan
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand. .,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand. .,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
44
|
Brenhouse HC, Schwarz JM. Immunoadolescence: Neuroimmune development and adolescent behavior. Neurosci Biobehav Rev 2016; 70:288-299. [PMID: 27260127 PMCID: PMC5412135 DOI: 10.1016/j.neubiorev.2016.05.035] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022]
Abstract
The brain is increasingly appreciated to be a constantly rewired organ that yields age-specific behaviors and responses to the environment. Adolescence in particular is a unique period characterized by continued brain maturation, superimposed with transient needs of the organism to traverse a leap from parental dependence to independence. Here we describe how these needs require immune maturation, as well as brain maturation. Our immune system, which protects us from pathogens and regulates inflammation, is in constant communication with our nervous system. Together, neuro-immune signaling regulates our behavioral responses to the environment, making this interaction a likely substrate for adolescent development. We review here the identified as well as understudied components of neuro-immune interactions during adolescence. Synaptic pruning, neurite outgrowth, and neurotransmitter release during adolescence all regulate-and are regulated by-immune signals, which occur via blood-brain barrier dynamics and glial activity. We discuss these processes, as well as how immune signaling during this transitional period of development confers differential effects on behavior and vulnerability to mental illness.
Collapse
Affiliation(s)
- Heather C Brenhouse
- Northeastern University, Psychology Department, 125 Nightingale Hall, Boston, MA 02115, United States.
| | - Jaclyn M Schwarz
- University of Delaware, Department of Psychological and Brain Sciences, 108 Wolf Hall, Newark, DE 19716, United States
| |
Collapse
|
45
|
Peterson LW, Philip NH, Dillon CP, Bertin J, Gough PJ, Green DR, Brodsky IE. Cell-Extrinsic TNF Collaborates with TRIF Signaling To Promote Yersinia-Induced Apoptosis. THE JOURNAL OF IMMUNOLOGY 2016; 197:4110-4117. [PMID: 27733552 DOI: 10.4049/jimmunol.1601294] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/20/2016] [Indexed: 02/02/2023]
Abstract
Innate immune responses that are crucial for control of infection are often targeted by microbial pathogens. Blockade of NF-κB and MAPK signaling by the Yersinia virulence factor YopJ inhibits cytokine production by innate immune cells but also triggers cell death. This cell death requires RIPK1 kinase activity and caspase-8, which are engaged by TLR4 and the adaptor protein TRIF. Nevertheless, TLR4- and TRIF-deficient cells undergo significant apoptosis, implicating TLR4/TRIF-independent pathways in the death of Yersinia-infected cells. In this article, we report a key role for TNF/TNFR1 in Yersinia-induced cell death of murine macrophages, which occurs despite the blockade of NF-κB and MAPK signaling imposed by Yersinia on infected cells. Intriguingly, direct analysis of YopJ injection revealed a heterogeneous population of injection-high and injection-low cells, and demonstrated that TNF expression came from the injection-low population. Moreover, TNF production by this subpopulation was necessary for maximal apoptosis in the population of highly injected cells, and TNFR-deficient mice displayed enhanced susceptibility to Yersinia infection. These data demonstrate an important role for collaboration between TNF and pattern recognition receptor signals in promoting maximal apoptosis during bacterial infection, and demonstrate that heterogeneity in virulence factor injection and cellular responses play an important role in promoting anti-Yersinia immune defense.
Collapse
Affiliation(s)
- Lance W Peterson
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104.,Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Naomi H Philip
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104.,Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Christopher P Dillon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105; and
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19422
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19422
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105; and
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104; .,Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
46
|
Inhibition of ROS and upregulation of inflammatory cytokines by FoxO3a promotes survival against Salmonella typhimurium. Nat Commun 2016; 7:12748. [PMID: 27599659 PMCID: PMC5023958 DOI: 10.1038/ncomms12748] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
Virulent intracellular pathogens, such as the Salmonella species, engage numerous virulence factors to subvert host defence mechanisms to induce a chronic infection that leads to typhoid or exacerbation of other chronic inflammatory conditions. Here we show the role of the forkhead transcription factor FoxO3a during infection of mice with Salmonella typhimurium (ST). Although FoxO3a signalling does not affect the development of CD8+ T cell responses to ST, FoxO3a has an important protective role, particularly during the chronic stage of infection, by limiting the persistence of oxidative stress. Furthermore, FoxO3a signalling regulates ERK signalling in macrophages, which results in the maintenance of a proinflammatory state. FoxO3a signalling does not affect cell proliferation or cell death. Thus, these results reveal mechanisms by which FoxO3a promotes host survival during infection with chronic, virulent intracellular bacteria. FoxO3a signalling has limited influence over acute bacterial infection. Here the authors show that FoxO3a promotes survival of mice in response to chronic Salmonella typhimurium infection by restraining oxidative stress and ERK signalling.
Collapse
|
47
|
Garib FY, Rizopulu AP. T-Regulatory Cells as Part of Strategy of Immune Evasion by Pathogens. BIOCHEMISTRY (MOSCOW) 2016; 80:957-71. [PMID: 26547064 DOI: 10.1134/s0006297915080015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Under physiological conditions, regulatory processes can suppress the immune response after elimination of a pathogen and restore homeostasis through the destruction and suppression of obsolete effector cells of the immune system. The main players in this process are T-regulatory cells (Tregs) and immature dendritic cells, which suppress the immune response by their own products and/or by inducing synthesis of immunosuppressive interleukins IL-10, IL-35, and transforming growth factor (TGF-β) by other cells. This mechanism is also used by widespread "successful" pathogens that are capable of chronically persisting in the human body - herpes virus, hepatitis viruses, human immunodeficiency virus, Mycobacterium tuberculosis, Helicobacter pylori, and others. During coevolution of microbial pathogens and the host immune system, the pathogens developed sophisticated strategies for evading the host defense, so-called immune evasion. In particular, molecular structures of pathogens during the interaction with dendritic cells via activating and inhibitory receptors can change intracellular signal transduction, resulting in block of maturation of dendritic cells. Immature dendritic cells become tolerogenic and cause differentiation of Tregs from the conventional T-cell CD4+. Microbial molecules can also react directly with Tregs through innate immune receptors. Costimulation of Toll-like receptor 5 (TLR5) by flagellin increases the expression of the transcription factor Foxp3, which increases the suppressive activity of Treg cells. From all evasion mechanisms, the induction of immunosuppression by Treg through IL-10, IL-35, and TGF-β appears most effective. This results in the suppression of inflammation and of adaptive immune responses against pathogens, optimizing the conditions for the survival of bacteria and viruses.
Collapse
Affiliation(s)
- F Yu Garib
- Lomonosov Moscow State University, Biological Faculty, Moscow, 119991, Russia
| | | |
Collapse
|
48
|
Outrunning the Red Queen: bystander activation as a means of outpacing innate immune subversion by intracellular pathogens. Cell Mol Immunol 2016; 14:14-21. [PMID: 27545071 PMCID: PMC5214943 DOI: 10.1038/cmi.2016.36] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 12/15/2022] Open
Abstract
Originally described by the late evolutionary biologist Leigh Van Valen, the Red Queen hypothesis posits that the evolutionary arms race between hosts and their pathogens selects for discrete, genetically encoded events that lead to competitive advantages over the other species. Examples of immune evasion strategies are seen throughout the co-evolution of the mammalian immune system and pathogens, such as the enzymatic inactivation of nuclear factor-κB signaling or host translation by pathogen-encoded virulence factors. Such immunoevasive maneuvers would be expected to select for the evolution of innate immune counterstrategies. Recent advances in our understanding of host immunity and microbial pathogenesis have provided insight into a particular innate immune adaptation, termed bystander activation. Bystander activation occurs as a consequence of infected cells alerting and instructing neighboring uninfected cells to produce inflammatory mediators, either through direct cell contact or paracrine signals. Thus, bystander activation can allow the immune system to overcome the ability of pathogens to disarm immune signaling in directly infected cells. This review presents an overview of the general hallmarks of bystander activation and their emerging role in innate immunity to intracellular pathogens, as well as examples of recent mechanistic discoveries relating to the bystander activation during infection with specific pathogens relevant to human health and disease.
Collapse
|
49
|
Wu CY, Hsu WL, Wang CH, Liang JL, Tsai MH, Yen CJ, Li HW, Chiu SJ, Chang CH, Huang YB, Lin MW, Yoshioka T. A Novel Strategy for TNF-Alpha Production by 2-APB Induced Downregulated SOCE and Upregulated HSP70 in O. tsutsugamushi-Infected Human Macrophages. PLoS One 2016; 11:e0159299. [PMID: 27472555 PMCID: PMC4966960 DOI: 10.1371/journal.pone.0159299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/30/2016] [Indexed: 12/20/2022] Open
Abstract
Orientia (O.) tsutsugamushi-induced scrub typhus is endemic across many regions of Asia and the Western Pacific, where an estimated 1 million cases occur each year; the majority of patients infected with O. tsutsugamushi end up with a cytokine storm from a severe inflammatory response. Previous reports have indicated that blocking tumor necrosis factor (TNF)-α reduced cell injury from a cytokine storm. Since TNF-α production is known to be associated with intracellular Ca2+ elevation, we examined the effect of store-operated Ca2+ entry (SOCE) inhibitors on TNF-α production in O. tsutsugamushi-infected macrophages. We found that 2-aminoethoxydiphenyl borate (2-APB), but not SKF96365, facilitates the suppression of Ca2+ mobilization via the interruption of Orai1 expression in O. tsutsugamushi-infected macrophages. Due to the decrease of Ca2+ elevation, the expression of TNF-α and its release from macrophages was repressed by 2-APB. In addition, a novel role of 2-APB was found in macrophages that causes the upregulation of heat shock protein 70 (HSP70) expression associated with ERK activation; upregulated TNF-α production in the case of knockdown HSP70 was inhibited with 2-APB treatment. Furthermore, elevated HSP70 formation unexpectedly did not help the cell survival of O. tsutsugamushi-infected macrophages. In conclusion, the parallelism between downregulated Ca2+ mobilization via SOCE and upregulated HSP70 after treatment with 2-APB against TNF-α production was found to efficiently attenuate an O. tsutsugamushi-induced severe inflammatory response.
Collapse
Affiliation(s)
- Ching-Ying Wu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Dermatology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Wen-Li Hsu
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | - Ming-Hsien Tsai
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Jung Yen
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiu-Wen Li
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Siou-Jin Chiu
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| | - Chung-Hsing Chang
- Department of Dermatology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yaw-Bin Huang
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Wei Lin
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (TY); (MWL)
| | - Tohru Yoshioka
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (TY); (MWL)
| |
Collapse
|
50
|
Long-Term Evolution of Burkholderia multivorans during a Chronic Cystic Fibrosis Infection Reveals Shifting Forces of Selection. mSystems 2016; 1:mSystems00029-16. [PMID: 27822534 PMCID: PMC5069766 DOI: 10.1128/msystems.00029-16] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/25/2016] [Indexed: 11/20/2022] Open
Abstract
Burkholderia multivorans is an opportunistic pathogen capable of causing severe disease in patients with cystic fibrosis (CF). Patients may be chronically infected for years, during which the bacterial population evolves in response to unknown forces. Here we analyze the genomic and functional evolution of a B. multivorans infection that was sequentially sampled from a CF patient over 20 years. The population diversified into at least four primary, coexisting clades with distinct evolutionary dynamics. The average substitution rate was only 2.4 mutations/year, but notably, some lineages evolved more slowly, whereas one diversified more rapidly by mostly nonsynonymous mutations. Ten loci, mostly involved in gene expression regulation and lipid metabolism, acquired three or more independent mutations and define likely targets of selection. Further, a broad range of phenotypes changed in association with the evolved mutations; they included antimicrobial resistance, biofilm regulation, and the presentation of lipopolysaccharide O-antigen repeats, which was directly caused by evolved mutations. Additionally, early isolates acquired mutations in genes involved in cyclic di-GMP (c-di-GMP) metabolism that associated with increased c-di-GMP intracellular levels. Accordingly, these isolates showed lower motility and increased biofilm formation and adhesion to CFBE41o- epithelial cells than the initial isolate, and each of these phenotypes is an important trait for bacterial persistence. The timing of the emergence of this clade of more adherent genotypes correlated with the period of greatest decline in the patient's lung function. All together, our observations suggest that selection on B. multivorans populations during long-term colonization of CF patient lungs either directly or indirectly targets adherence, metabolism, and changes in the cell envelope related to adaptation to the biofilm lifestyle. IMPORTANCE Bacteria may become genetically and phenotypically diverse during long-term colonization of cystic fibrosis (CF) patient lungs, yet our understanding of within-host evolutionary processes during these infections is lacking. Here we combined current genome sequencing technologies and detailed phenotypic profiling of the opportunistic pathogen Burkholderia multivorans using sequential isolates sampled from a CF patient over 20 years. The evolutionary history of these isolates highlighted bacterial genes and pathways that were likely subject to strong selection within the host and were associated with altered phenotypes, such as biofilm production, motility, and antimicrobial resistance. Importantly, multiple lineages coexisted for years or even decades within the infection, and the period of diversification within the dominant lineage was associated with deterioration of the patient's lung function. Identifying traits under strong selection during chronic infection not only sheds new light onto Burkholderia evolution but also sets the stage for tailored therapeutics targeting the prevailing lineages associated with disease progression.
Collapse
|