1
|
Andrieu J, Mège J, Mezouar S. Monkeypox Virus and Pregnancy. J Med Virol 2025; 97:e70337. [PMID: 40223710 PMCID: PMC11995370 DOI: 10.1002/jmv.70337] [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: 01/27/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Human monkeypox (Mpox) is a zoonotic disease caused by monkeypox virus (MPXV) present in western Africa and exported sporadically worldwide. MPXV causes illness in individuals and pregnant women which constitute a population at risk with obstetrical and fetal complications including miscarriage, stillbirth and premature delivery. There are accumulated data suggesting a vertical transmission of MPXV from mother to fetus. This review provides an overview of the literature on MPXV infection in pregnant women with a specific focus on vertical transmission.
Collapse
Affiliation(s)
- Jonatane Andrieu
- Aix‐Marseille Univ, Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio‐culturelle, Droit, Éthique et SantéMarseilleFrance
| | - Jean‐louis Mège
- Aix‐Marseille Univ, Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio‐culturelle, Droit, Éthique et SantéMarseilleFrance
- Department of ImmunologyTimone HospitalMarseilleFrance
| | - Soraya Mezouar
- Aix‐Marseille Univ, Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio‐culturelle, Droit, Éthique et SantéMarseilleFrance
- Faculty of Medical and Paramedical SciencesAix‐Marseille University, HIPE Human LabMarseilleFrance
| |
Collapse
|
2
|
Steinmetz AR, Pierce M, Martini A, Tholomier C, Manyam G, Chen Y, Sood A, Duplisea JJ, Johnson BA, Czerniak BA, Lee BH, Jagannath C, Yla-Herttuala S, Parker NR, McConkey DJ, Dinney CP, Mokkapati S. Single-cell RNA sequencing analysis identifies acute changes in the tumor microenvironment induced by interferon α gene therapy in a murine bladder cancer model. Front Immunol 2024; 15:1387229. [PMID: 39559365 PMCID: PMC11570268 DOI: 10.3389/fimmu.2024.1387229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Nadofaragene firadenovec (Ad-IFNα/Syn3) is now approved for BCG-unresponsive bladder cancer (BLCA). IFNα is a pleiotropic cytokine that causes direct tumor cell killing via TRAIL-mediated apoptosis, angiogenesis inhibition, and activation of the innate and adaptive immune system. We established an immunocompetent murine BLCA model to study the effects of murine adenoviral IFNα (muAd-Ifnα) gene therapy on cancer cells and the tumor microenvironment using a novel murine equivalent of Nadofaragene firadenovec (muAd-Ifnα). Methods Tumors were induced by instilling MB49 cells into the bladders of mice; luciferase imaging confirmed tumor development. Mice were treated with adenovirus control (Ad-Ctrl; empty vector), or muAd-Ifnα (3x1011 VP/mL), and survival analysis was performed. For single-cell sequencing (scRNAseq) analysis (72h), bladders were harvested and treated with collagenase/hyaluronidase and TrypLE for cell dissociation. Single cells were suspended in PBS/1% FBS buffer; viability was assessed with Vicell cell counter. scRNAseq analysis was performed using 10X genomics 3' sequencing. Raw RNAseq data were pre-processed using Cell Ranger single-cell software. Seurat (R package) was used to normalize and cluster the scRNA data. Pooled differential gene expression analysis in specific cell clusters was performed with DESeq2. Results We identified 16 cell clusters based on marker expression which were grouped into epithelial (tumor), uroplakin-enriched, endothelial, T-cells, neutrophils, and macrophage clusters. Top differentially expressed genes between muAd-Ifnα and Ad-Ctrl were identified. Within the specific cell clusters, IPA analysis revealed significant differences between muAd-Ifnα and control. IFNα signaling and hypercytokinemia/chemokinemia were upregulated in all clusters. Cell death pathways were upregulated in tumor and endothelial clusters. T-cells demonstrated upregulation of the immunogenic cell death signaling pathway and a decrease in the Th2 pathway genes. Macrophages showed upregulation of PD1/PD-L1 pathways along with downregulation of macrophage activation pathways (alternate and classical). Multiplex immunofluorescence confirmed increased infiltration with macrophages in muAd-Ifnα treated tumors compared to controls. PD1/PD-L1 expression was reduced at 72h. Discussion This single-cell analysis builds upon our understanding of the impact of Ad-IFNα on tumor cells and other compartments of the microenvironment. These data will help identify mechanisms to improve patient selection and therapeutic efficacy of Nadofaragene firadenovec.
Collapse
Affiliation(s)
- Alexis R. Steinmetz
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Morgan Pierce
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alberto Martini
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Come Tholomier
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ganiraju Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yan Chen
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Akshay Sood
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jonathan J. Duplisea
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Burles A. Johnson
- Johns Hopkins Greenberg Bladder Cancer Institute, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Bogdan A. Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Byron H. Lee
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | | | - Nigel R. Parker
- A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - David J. McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Colin P. Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sharada Mokkapati
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
3
|
Hu Z, Li Y, Yang J, Liu J, Zhou H, Sun C, Tian C, Zhu C, Shao M, Wang S, Wei L, Liu M, Li S, Wang J, Xu H, Zhu W, Li X, Li J. Improved antitumor effectiveness of oncolytic HSV-1 viruses engineered with IL-15/IL-15Rα complex combined with oncolytic HSV-1-aPD1 targets colon cancer. Sci Rep 2024; 14:23671. [PMID: 39389985 PMCID: PMC11467195 DOI: 10.1038/s41598-024-72888-w] [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: 02/28/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Oncolytic virotherapy is emerging as a promising therapeutic avenue for cancer treatment, harnessing both innate and tumor-specific immune responses for targeted tumor elimination. In this study, we present a novel oncolytic virus (oHSV1-IL15B) derived from herpes simplex virus-1 (HSV-1), armed with IL-15/IL-15Rα complex, with a focus on treating colon cancer combined with oncolytic HSV-1 expressing anti-PD-1 antibody (oHSV1-aPD1). Results from our study reveal that recombinant oHSV-1 virus equipped with IL-15/IL-15Rα complex exhibited significant anti-tumor effects in a murine CT26 colon adenocarcinoma model. Notably, oHSV1-IL15B combined with oHSV-1-aPD1 demonstrates superior tumor inhibition and prolonged overall survival compared to oHSV1-mock and monotherapy groups. Further exploration highlights the impact of oHSV1-IL15B, oHSV-1-aPD1 and combined group on antitumor capacity, revealing a substantial increase in CD8+ T and CD4+ T cell proportions of CT26-bearing BALB/c mice and promoting apoptosis in tumor tissue. The study emphasizes the pivotal role of cytotoxic CD8+T cells in oncolytic virotherapy, demonstrating that recombinant oHSV1-IL15B combined with oncolytic HSV-1-aPD1 induces a robust tumor-specific T cell response. RNA sequence analysis highlighted oHSV1-IL15B combined with oHSV1-aPD1 improved tumors immune microenvironment on immune response, antiviral response-related genes and apoptosis-related genes, which contributed to anti-tumor immunotherapy. The findings underscore the promising antitumor activity achieved through the combination of IL-15/IL-15Rα complex and anti-PD-1 antibody with oHSV-1. This research opens avenues for diverse therapeutic strategies, suggesting the potential of synergistically utilizing cytokines and anti-PD-1 antibody with oncolytic viruses to enhance immunotherapy for cancer management.
Collapse
Affiliation(s)
- Zongfeng Hu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Yixiao Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | | | - Jiajia Liu
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Hua Zhou
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chunyang Sun
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chao Tian
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chengyang Zhu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Mingxia Shao
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Shengrun Wang
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Lijun Wei
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Min Liu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Shuzhen Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Jinyu Wang
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Haitian Xu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Wei Zhu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Xiaopeng Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China.
- Beijing WellGene Company, Ltd, Beijing, 100085, China.
| | - Jingfeng Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China.
| |
Collapse
|
4
|
Salerno-Goncalves R, Chen H, Bafford AC, Sztein MB. Epigenetic regulation in epithelial cells and innate lymphocyte responses to S. Typhi infection: insights into IFN-γ production and intestinal immunity. Front Immunol 2024; 15:1448717. [PMID: 39372404 PMCID: PMC11450450 DOI: 10.3389/fimmu.2024.1448717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/08/2024] [Indexed: 10/08/2024] Open
Abstract
Infection by Salmonella enterica serovar Typhi (S. Typhi), the cause of enteric fevers, is low in high-income countries but persistent in low- and middle-income countries, resulting in 65,400-187,700 deaths yearly. Drug resistance, including in the United States, exacerbates this issue. Evidence indicates that innate lymphocytes (INLs), such as natural killer (NK) cells, and unconventional T lymphocytes (e.g., Mucosal-associated invariant T (MAIT) cells and T-cell receptor gamma delta (TCR-γδ) cells) can impact the intestinal epithelial barrier, the primary site of exposure to S. Typhi. Moreover, INL production of IFN-γ is central in controlling S. Typhi infection. However, the impact of epithelial cells (EC) on the secretion of IFN-γ by INLs and the relationship between these events and epigenetic changes remains unknown. Epigenetic modifications in host cells are fundamental for their differentiation and function, including IFN-γ production. Herein, using a human organoid-derived polarized intestinal epithelial cell monolayer, we investigated the role of H3K4me3 and H3K27me3 epigenetic marks in intestinal immunity, focusing on the function of EC, NK, MAIT, and TCR-γδ cells in response to S. Typhi. This study builds on our previous findings that MAIT subsets exhibiting specific IFN-γ pattern signatures were associated with protection against typhoid fever and that S. Typhi infection regulates changes in chromatin marks that depend on individual cell subsets. Here, we show that cultures exposed to S. Typhi without EC exhibit a significant increase in NK and MAIT cells, and, to a lesser extent, TCR-γδ cells, expressing IFN-γ and H3K4me3 but not H3K27me3 marks, contrasting with cultures where EC is present. The influence of EC on INL H3K4me3 marks might be indirectly mediated through the modulation of IL-18 secretion via the Histone Deacetylase 6 gene during S. Typhi infection.
Collapse
Affiliation(s)
- Rosângela Salerno-Goncalves
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Haiyan Chen
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bafford
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Oncology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
5
|
Sang L, Gong X, Huang Y, Zhang L, Sun J. Immunotherapeutic implications on targeting the cytokines produced in rhinovirus-induced immunoreactions. FRONTIERS IN ALLERGY 2024; 5:1427762. [PMID: 38859875 PMCID: PMC11163110 DOI: 10.3389/falgy.2024.1427762] [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: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Rhinovirus is a widespread virus associated with several respiratory diseases, especially asthma exacerbation. Currently, there are no accurate therapies for rhinovirus. Encouragingly, it is found that during rhinovirus-induced immunoreactions the levels of certain cytokines in patients' serum will alter. These cytokines may have pivotal pro-inflammatory or anti-inflammatory effects via their specific mechanisms. Thus far, studies have shown that inhibitions of cytokines such as IL-1, IL-4, IL-5, IL-6, IL-13, IL-18, IL-25, and IL-33 may attenuate rhinovirus-induced immunoreactions, thereby relieving rhinovirus infection. Furthermore, such therapeutics for rhinovirus infection can be applied to viruses of other species, with certain practicability.
Collapse
Affiliation(s)
- Le Sang
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Xia Gong
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Yunlei Huang
- Department of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province, China
| | - Linling Zhang
- Department of Respiratory Medicine, Shaoxing People’s Hospital, Shaoxing City, Zhejiang Province, China
| | - Jian Sun
- Department of Respiratory Medicine, Shaoxing People’s Hospital, Shaoxing City, Zhejiang Province, China
| |
Collapse
|
6
|
Lepretre F, Gras D, Chanez P, Duez C. Natural killer cells in the lung: potential role in asthma and virus-induced exacerbation? Eur Respir Rev 2023; 32:230036. [PMID: 37437915 DOI: 10.1183/16000617.0036-2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/23/2023] [Indexed: 07/14/2023] Open
Abstract
Asthma is a chronic inflammatory airway disorder whose pathophysiological and immunological mechanisms are not completely understood. Asthma exacerbations are mostly driven by respiratory viral infections and characterised by worsening of symptoms. Despite current therapies, asthma exacerbations can still be life-threatening. Natural killer (NK) cells are innate lymphoid cells well known for their antiviral activity and are present in the lung as circulating and resident cells. However, their functions in asthma and its exacerbations are still unclear. In this review, we will address NK cell activation and functions, which are particularly relevant for asthma and virus-induced asthma exacerbations. Then, the role of NK cells in the lungs at homeostasis in healthy individuals will be described, as well as their functions during pulmonary viral infections, with an emphasis on those associated with asthma exacerbations. Finally, we will discuss the involvement of NK cells in asthma and virus-induced exacerbations and examine the effect of asthma treatments on NK cells.
Collapse
Affiliation(s)
- Florian Lepretre
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Delphine Gras
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Pascal Chanez
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- APHM, Hôpital Nord, Clinique des Bronches, de l'allergie et du sommeil, Marseille, France
| | - Catherine Duez
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| |
Collapse
|
7
|
Wang R, Cui W, Yang H. The interplay between innate lymphoid cells and microbiota. mBio 2023; 14:e0039923. [PMID: 37318214 PMCID: PMC10470585 DOI: 10.1128/mbio.00399-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/21/2023] [Indexed: 06/16/2023] Open
Abstract
Innate lymphoid cells (ILCs) are mainly resident in mucosal tissues such as gastrointestinal tract and respiratory tract, so they are closely linked to the microbiota. ILCs can protect commensals to maintain homeostasis and increase resistance to pathogens. Moreover, ILCs also play an early role in defense against a variety of pathogenic microorganisms including pathogenic bacteria, viruses, fungi and parasites, before the intervention of adaptive immune system. Due to the lack of adaptive antigen receptors expressed on T cells and B cells, ILCs need to use other means to sense the signals of microbiota and play a role in corresponding regulation. In this review, we focus on and summarize three major mechanisms used in the interaction between ILCs and microbiota: the mediation of accessory cells represented by dendritic cells; the metabolic pathways of microbiota or diet; the participation of adaptive immune cells.
Collapse
Affiliation(s)
- Rui Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Wenwen Cui
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
8
|
Qudus MS, Cui X, Tian M, Afaq U, Sajid M, Qureshi S, Liu S, Ma J, Wang G, Faraz M, Sadia H, Wu K, Zhu C. The prospective outcome of the monkeypox outbreak in 2022 and characterization of monkeypox disease immunobiology. Front Cell Infect Microbiol 2023; 13:1196699. [PMID: 37533932 PMCID: PMC10391643 DOI: 10.3389/fcimb.2023.1196699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/21/2023] [Indexed: 08/04/2023] Open
Abstract
A new threat to global health re-emerged with monkeypox's advent in early 2022. As of November 10, 2022, nearly 80,000 confirmed cases had been reported worldwide, with most of them coming from places where the disease is not common. There were 53 fatalities, with 40 occurring in areas that had never before recorded monkeypox and the remaining 13 appearing in the regions that had previously reported the disease. Preliminary genetic data suggest that the 2022 monkeypox virus is part of the West African clade; the virus can be transmitted from person to person through direct interaction with lesions during sexual activity. It is still unknown if monkeypox can be transmitted via sexual contact or, more particularly, through infected body fluids. This most recent epidemic's reservoir host, or principal carrier, is still a mystery. Rodents found in Africa can be the possible intermediate host. Instead, the CDC has confirmed that there are currently no particular treatments for monkeypox virus infection in 2022; however, antivirals already in the market that are successful against smallpox may mitigate the spread of monkeypox. To protect against the disease, the JYNNEOS (Imvamune or Imvanex) smallpox vaccine can be given. The spread of monkeypox can be slowed through measures such as post-exposure immunization, contact tracing, and improved case diagnosis and isolation. Final Thoughts: The latest monkeypox epidemic is a new hazard during the COVID-19 epidemic. The prevailing condition of the monkeypox epidemic along with coinfection with COVID-19 could pose a serious condition for clinicians that could lead to the global epidemic community in the form of coinfection.
Collapse
Affiliation(s)
- Muhammad Suhaib Qudus
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xianghua Cui
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Uzair Afaq
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Muhammad Sajid
- RNA Therapeutics Institute, Chan Medical School, University of Massachusetts Worcester, Worcester, MA, United States
| | - Sonia Qureshi
- Krembil Research Institute, University of Health Network, Toronto, ON, Canada
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - June Ma
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guolei Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Muhammad Faraz
- Department of Microbiology, Quaid-I- Azam University, Islamabad, Pakistan
| | - Haleema Sadia
- Department of Biotechnology, Baluchistan University of Information Technology, Engineering and Management Sciences (BUITEMS), Quetta, Pakistan
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
9
|
Moioffer SJ, Berton RR, McGonagill PW, Jensen IJ, Griffith TS, Badovinac VP. Inefficient Recovery of Repeatedly Stimulated Memory CD8 T Cells after Polymicrobial Sepsis Induction Leads to Changes in Memory CD8 T Cell Pool Composition. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:168-179. [PMID: 36480268 PMCID: PMC9840817 DOI: 10.4049/jimmunol.2200676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/09/2022] [Indexed: 01/03/2023]
Abstract
Long-lasting sepsis-induced immunoparalysis has been principally studied in primary (1°) memory CD8 T cells; however, the impact of sepsis on memory CD8 T cells with a history of repeated cognate Ag encounters is largely unknown but important in understanding the role of sepsis in shaping the pre-existing memory CD8 T cell compartment. Higher-order memory CD8 T cells are crucial in providing immunity against common pathogens that reinfect the host or are generated by repeated vaccination. In this study, we analyzed peripheral blood from septic patients and show that memory CD8 T cells with defined Ag specificity for recurring CMV infection proliferate less than bulk populations of central memory CD8 T cells. Using TCR-transgenic T cells to generate 1° and higher-order (quaternary [4°]) memory T cells within the same host, we demonstrate that the susceptibility and loss of both memory subsets are similar after sepsis induction, and sepsis diminished Ag-dependent and -independent (bystander) functions of these memory subsets equally. Both the 1° and 4° memory T cell populations proliferated in a sepsis-induced lymphopenic environment; however, due to the intrinsic differences in baseline proliferative capacity, expression of receptors (e.g., CD127/CD122), and responsiveness to homeostatic cytokines, 1° memory T cells become overrepresented over time in sepsis survivors. Finally, IL-7/anti-IL-7 mAb complex treatment early after sepsis induction preferentially rescued the proliferation and accumulation of 1° memory T cells, whereas recovery of 4° memory T cells was less pronounced. Thus, inefficient recovery of repeatedly stimulated memory cells after polymicrobial sepsis induction leads to changes in memory T cell pool composition, a notion with important implications in devising strategies to recover the number and function of pre-existing memory CD8 T cells in sepsis survivors.
Collapse
Affiliation(s)
| | - Roger R. Berton
- Department of Pathology, University of Iowa, Iowa City, IA;,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | | | - Isaac J. Jensen
- Columbia University Irving Medical Center, University of Minnesota, Minneapolis, MN
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN,,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA;,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| |
Collapse
|
10
|
Mukherjee AG, Wanjari UR, Kannampuzha S, Das S, Murali R, Namachivayam A, Renu K, Ramanathan G, Doss C GP, Vellingiri B, Dey A, Valsala Gopalakrishnan A. The pathophysiological and immunological background of the monkeypox virus infection: An update. J Med Virol 2023; 95:e28206. [PMID: 36217803 DOI: 10.1002/jmv.28206] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 01/18/2023]
Abstract
In addition to the COVID-19 waves, the globe is facing global monkeypox (MPX) outbreak. MPX is an uncommon zoonotic infection characterized by symptoms similar to smallpox. It is caused by the monkeypox virus (MPXV), a double-stranded DNA virus that belongs to the genus Orthopoxvirus (OPXV). MPXV, which causes human disease, has been confined to Africa for many years, with only a few isolated cases in other areas. Outside of Africa, the continuing MPXV outbreak in multiple countries in 2022 is the greatest in recorded history. The current outbreak, with over 10 000 confirmed cases in over 50 countries between May and July 2022, demonstrates that MPXV may travel rapidly among humans and pose a danger to human health worldwide. The rapid spread of such outbreaks in recent times has elevated MPX to the status of a rising zoonotic disease with significant epidemic potential. While the MPXV is not as deadly or contagious as the variola virus that causes smallpox, it poses a threat because it could evolve into a more potent human pathogen. This review assesses the potential threat to the human population and provides a brief overview of what is currently known about this reemerging virus. By analyzing the biological effects of MPXV on human health, its shifting epidemiological footprint, and currently available therapeutic options, this review has presented the most recent insights into the biology of the virus. This study also clarifies the key potential causes that could be to blame for the present MPX outbreak and draw attention to major research questions and promising new avenues for combating the current MPX epidemic.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Soumik Das
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Kaviyarasi Renu
- Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - George Priya Doss C
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Balachandar Vellingiri
- Department of Human Genetics and Molecular Biology, Human Molecular Cytogenetics and Stem Cell Laboratory, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
11
|
Muehling LM, Heymann PW, Carper H, Murphy DD, Rajadhyaksha E, Kennedy J, Early SV, Soto‐Quiros M, Avila L, Workman L, Platts‐Mills TAE, Woodfolk JA. Cluster analysis of nasal cytokines during rhinovirus infection identifies different immunophenotypes in both children and adults with allergic asthma. Clin Exp Allergy 2022; 52:1169-1182. [PMID: 35575980 PMCID: PMC9547929 DOI: 10.1111/cea.14176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Infection with rhinovirus (RV) is a major risk factor for disease exacerbations in patients with allergic asthma. This study analysed a broad set of cytokines in the noses of children and adults with asthma during RV infection in order to identify immunophenotypes that may link to virus-induced episodes. METHODS Nasal wash specimens were analysed in children (n = 279 [healthy, n = 125; stable asthma, n = 64; wheeze, n = 90], ages 2-12) who presented to a hospital emergency department, and in adults (n = 44 [healthy, n = 13; asthma, n = 31], ages 18-38) who were experimentally infected with RV, including a subset who received anti-IgE. Cytokines were measured by multiplex bead assay and data analysed by univariate and multivariate methods to test relationships to viral load, allergic status, airway inflammation, and clinical outcomes. RESULTS Analysis of a core set of 7 cytokines (IL-6, CXCL8/IL-8, IL-15, EGF, G-CSF, CXCL10/IP-10 and CCL22/MDC) revealed higher levels in children with acute wheeze versus those with stable asthma or controls. Multivariate analysis identified two clusters that were enriched for acutely wheezing children; one displaying high viral load ("RV-high") with robust secretion of CXCL10, and the other displaying high IgE with elevated EGF, CXCL8 and both eosinophil- and neutrophil-derived mediators. Broader assessment of 39 cytokines confirmed that children with acute wheeze were not deficient in type 1 anti-viral responses. Analysis of 18 nasal cytokines in adults with asthma who received RV challenge identified two clusters; one that was "RV-high" and linked to robust induction of anti-viral cytokines and anti-IgE; and the other associated with more severe symptoms and a higher inflammatory state featuring eosinophil and neutrophil factors. CONCLUSIONS The results confirm the presence of different immunophenotypes linked to parameters of airway disease in both children and adults with asthma who are infected with RV. Such discrepancies may reflect the ability to regulate anti-viral responses.
Collapse
Affiliation(s)
- Lyndsey M. Muehling
- Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Peter W. Heymann
- Department of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Holliday Carper
- Department of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Deborah D. Murphy
- Department of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Evan Rajadhyaksha
- Department of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Joshua Kennedy
- Department of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA,Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Stephen V. Early
- Department of Otolaryngology‐Head and Neck SurgeryUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | | | | | - Lisa Workman
- Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | | | - Judith A. Woodfolk
- Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| |
Collapse
|
12
|
Lum FM, Torres-Ruesta A, Tay MZ, Lin RTP, Lye DC, Rénia L, Ng LFP. Monkeypox: disease epidemiology, host immunity and clinical interventions. Nat Rev Immunol 2022; 22:597-613. [PMID: 36064780 PMCID: PMC9443635 DOI: 10.1038/s41577-022-00775-4] [Citation(s) in RCA: 253] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 12/11/2022]
Abstract
Monkeypox virus (MPXV), which causes disease in humans, has for many years been restricted to the African continent, with only a handful of sporadic cases in other parts of the world. However, unprecedented outbreaks of monkeypox in non-endemic regions have recently taken the world by surprise. In less than 4 months, the number of detected MPXV infections has soared to more than 48,000 cases, recording a total of 13 deaths. In this Review, we discuss the clinical, epidemiological and immunological features of MPXV infections. We also highlight important research questions and new opportunities to tackle the ongoing monkeypox outbreak.
Collapse
Affiliation(s)
- Fok-Moon Lum
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anthony Torres-Ruesta
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Matthew Z Tay
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Raymond T P Lin
- National Public Health Laboratory, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK.
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
13
|
Mikkola H, Honkila M, Tapiainen T, Jartti T. Susceptibility to rhinovirus-induced early wheezing as a risk factor for subsequent asthma development. CURRENT RESPIRATORY MEDICINE REVIEWS 2022. [DOI: 10.2174/1573398x18666220103113813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Rhinovirus is one of the two most common viral agents that cause bronchiolitis in young children. During the first 12 months, it is second to the respiratory syncytial virus, but after 12 months, it begins dominating the statistics. Wheezing and dry cough are typical clinical symptoms indicative of rhinovirus-induced bronchiolitis, although overlap of symptoms with other virus infections is common. Several studies have shown that atopic predisposition and reduced interferon responses increase susceptibility to rhinovirus-induced wheezing. More recent studies have found that certain genetic variations at strong asthma loci also increase susceptibility. Rhinovirus-induced wheezing in the early years of life is known to increase the risk of subsequent asthma development and may be associated with airway remodeling. This risk is increased by aeroallergen sensitization. Currently, there are no clinically approved preventive treatments for asthma. However, studies show promising results indicating that children with rhinovirus-affected first-time wheezing respond to bronchodilators in terms of less short-term symptoms and that controlling airway inflammatory responses with anti-inflammatory medication may markedly decrease asthma development. Also, enhancing resistance to respiratory viruses has been a topic of discussion. Primary and secondary prevention strategies are being developed with the aim of decreasing the incidence of asthma. Here, we review the current knowledge on rhinovirus-induced early wheezing as a risk factor for subsequent asthma development and related asthma-prevention strategies.
Collapse
Affiliation(s)
- Hannele Mikkola
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Minna Honkila
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Terhi Tapiainen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Tuomas Jartti
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
14
|
Sobkowiak MJ, Paquin-Proulx D, Bosnjak L, Moll M, Sällberg Chen M, Sandberg JK. Dynamics of IL-15/IL-15R-α expression in response to HSV-1 infection reveal a novel mode of viral immune evasion counteracted by iNKT cells. Eur J Immunol 2021; 52:462-471. [PMID: 34910820 DOI: 10.1002/eji.202149287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 10/12/2021] [Accepted: 12/10/2021] [Indexed: 11/10/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) infects and persists in most of the human population. Interleukin-15 (IL-15) has an important role in the activation of cell-mediated immune responses and acts in complex with IL-15 receptor alpha (IL-15R-α) through cell surface transpresentation. Here, we have examined the IL-15/IL-15R-α complex response dynamics during HSV-1 infection in human keratinocytes. Surface expression of the IL-15/IL-15R-α complex rapidly increased in response to HSV-1, reaching a peak around 12 h after infection. This response was dependent on detection of viral replication by TLR3, and enhancement of IL15 and IL15RA gene expression. Beyond the peak of expression, levels of IL-15 and IL-15R-α gradually declined, reaching a profound loss of surface expression beyond 24 h of infection. This involved the loss of IL15 and IL15RA transcription. Interestingly, invariant natural killer T (iNKT) cells inhibited the viral interference with IL-15/IL-15R-α complex expression in an IFNγ-dependent manner. These results indicate that rapid upregulation of the IL-15/IL-15R-α complex occurs in HSV-1 infected keratinocytes, and that this response is targeted by viral interference. Shutdown of the IL-15 axis represents a novel mode of HSV-1 immune evasion, which can be inhibited by the host iNKT cell response.
Collapse
Affiliation(s)
- Michał J Sobkowiak
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden.,Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Dominic Paquin-Proulx
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden
| | - Lidija Bosnjak
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden
| | - Markus Moll
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden
| | | | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Stockholm, Sweden
| |
Collapse
|
15
|
Krebs R, Tikkanen JM, Raissadati A, Hollmén M, Dhaygude K, Lemström KB. Inhibition of Vascular Endothelial Growth Factor Receptors 1 and 2 Attenuates Natural Killer Cell and Innate Immune Responses in an Experimental Model for Obliterative Bronchiolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 192:254-269. [PMID: 34774518 DOI: 10.1016/j.ajpath.2021.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023]
Abstract
Obliterative bronchiolitis (OB) after lung transplantation is a nonreversible, life-threatening complication. We investigated the role of vascular endothelial growth factor receptor (VEGFR)-1 and -2 in the development of obliterative airway disease (OAD), an experimental model for OB. The nonimmunosuppressed recipients underwent transplantation with fully major histocompatibility complex mismatched heterotopic tracheal allografts and received VEGFR-1 and -2-specific monoclonal antibodies either alone or in combination or rat IgG as a control. The treatment with VEGFR-1- or -2-blocking antibody significantly decreased intragraft mRNA expression of natural killer cell activation markers early after transplantation. This was followed by reduced infiltration of CD11b+ cells and CD4+ T cells as well as down-regulated mRNA expression of proinflammatory chemokines and profibrotic growth factors. However, blocking of both VEGFR-1 and -2 was necessary to reduce luminal occlusion. Furthermore, concomitant inhibition of the calcineurin activation pathway almost totally abolished the development of OAD. This study proposes that blocking of VEGF receptors blunted natural killer cell and innate immune responses early after transplantation and attenuated the development of OAD. The results of this study suggest that further studies on the role of VEGFR-1 and -2 blocking in development of obliterative airway lesions might be rewarding.
Collapse
Affiliation(s)
- Rainer Krebs
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland.
| | - Jussi M Tikkanen
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland; Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Alireza Raissadati
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Maria Hollmén
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Kishor Dhaygude
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Karl B Lemström
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland; Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Abstract
Pneumonia is a common acute respiratory infection that affects the alveoli and distal airways; it is a major health problem and associated with high morbidity and short-term and long-term mortality in all age groups worldwide. Pneumonia is broadly divided into community-acquired pneumonia or hospital-acquired pneumonia. A large variety of microorganisms can cause pneumonia, including bacteria, respiratory viruses and fungi, and there are great geographical variations in their prevalence. Pneumonia occurs more commonly in susceptible individuals, including children of <5 years of age and older adults with prior chronic conditions. Development of the disease largely depends on the host immune response, with pathogen characteristics having a less prominent role. Individuals with pneumonia often present with respiratory and systemic symptoms, and diagnosis is based on both clinical presentation and radiological findings. It is crucial to identify the causative pathogens, as delayed and inadequate antimicrobial therapy can lead to poor outcomes. New antibiotic and non-antibiotic therapies, in addition to rapid and accurate diagnostic tests that can detect pathogens and antibiotic resistance will improve the management of pneumonia.
Collapse
|
17
|
Williams TC, Jackson DJ, Maltby S, Walton RP, Ching YM, Glanville N, Singanayagam A, Brewins JJ, Clarke D, Hirsman AG, Loo SL, Wei L, Beale JE, Casolari P, Caramori G, Papi A, Belvisi M, Wark PAB, Johnston SL, Edwards MR, Bartlett NW. Rhinovirus-induced CCL17 and CCL22 in Asthma Exacerbations and Differential Regulation by STAT6. Am J Respir Cell Mol Biol 2021; 64:344-356. [PMID: 33264064 PMCID: PMC7909342 DOI: 10.1165/rcmb.2020-0011oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
The interplay of type-2 inflammation and antiviral immunity underpins asthma exacerbation pathogenesis. Virus infection induces type-2 inflammation-promoting chemokines CCL17 and CCL22 in asthma; however, mechanisms regulating induction are poorly understood. By using a human rhinovirus (RV) challenge model in human airway epithelial cells in vitro and mice in vivo, we assessed mechanisms regulating CCL17 and CCL22 expression. Subjects with mild to moderate asthma and healthy volunteers were experimentally infected with RV and airway CCL17 and CCL22 protein quantified. In vitro airway epithelial cell- and mouse-RV infection models were then used to define STAT6- and NF-κB-mediated regulation of CCL17 and CCL22 expression. Following RV infection, CCL17 and CCL22 expression was higher in asthma, which differentially correlated with clinical and immunological parameters. Air-liquid interface-differentiated primary epithelial cells from donors with asthma also expressed higher levels of RV-induced CCL22. RV infection boosted type-2 cytokine-induced STAT6 activation. In epithelial cells, type-2 cytokines and STAT6 activation had differential effects on chemokine expression, increasing CCL17 and suppressing CCL22, whereas NF-κB promoted expression of both chemokines. In mice, RV infection activated pulmonary STAT6, which was required for CCL17 but not CCL22 expression. STAT6-knockout mice infected with RV expressed increased levels of NF-κB-regulated chemokines, which was associated with rapid viral clearance. Therefore, RV-induced upregulation of CCL17 and CCL22 was mediated by NF-κB activation, whereas expression was differentially regulated by STAT6. Together, these findings suggest that therapeutic targeting of type-2 STAT6 activation alone will not block all inflammatory pathways during RV infection in asthma.
Collapse
Affiliation(s)
- Teresa C. Williams
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - David J. Jackson
- Asthma UK Centre, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Guy’s Severe Asthma Centre, Guy’s & St. Thomas’ National Health Service Trust, London, United Kingdom
| | - Steven Maltby
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Ross P. Walton
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yee-Mann Ching
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas Glanville
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jennifer J. Brewins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Deborah Clarke
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory, Inflammation and Autoimmunity Department, MedImmune, Cambridge, United Kingdom
| | - Aurica G. Hirsman
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Su-Ling Loo
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Lan Wei
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Janine E. Beale
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Paolo Casolari
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Gaetano Caramori
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
- Dipartimento di Scienze Biomediche, Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali, Università degli Studi di Messina, Messina, Italy; and
| | - Alberto Papi
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Maria Belvisi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory, Inflammation and Autoimmunity Department, MedImmune, Cambridge, United Kingdom
| | - Peter A. B. Wark
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | | | - Michael R. Edwards
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nathan W. Bartlett
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Michi AN, Love ME, Proud D. Rhinovirus-Induced Modulation of Epithelial Phenotype: Role in Asthma. Viruses 2020; 12:v12111328. [PMID: 33227953 PMCID: PMC7699223 DOI: 10.3390/v12111328] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Human rhinoviruses have been linked both to the susceptibility of asthma development and to the triggering of acute exacerbations. Given that the human airway epithelial cell is the primary site of human rhinovirus (HRV) infection and replication, the current review focuses on how HRV-induced modulation of several aspects of epithelial cell phenotype could contribute to the development of asthma or to the induction of exacerbations. Modification of epithelial proinflammatory and antiviral responses are considered, as are alterations in an epithelial barrier function and cell phenotype. The contributions of the epithelium to airway remodeling and to the potential modulation of immune responses are also considered. The potential interactions of each type of HRV-induced epithelial phenotypic changes with allergic sensitization and allergic phenotype are also considered in the context of asthma development and of acute exacerbations.
Collapse
|
19
|
van der Heide SL, Xi Y, Upham JW. Natural Killer Cells and Host Defense Against Human Rhinoviruses Is Partially Dependent on Type I IFN Signaling. Front Cell Infect Microbiol 2020; 10:510619. [PMID: 33194777 PMCID: PMC7609819 DOI: 10.3389/fcimb.2020.510619] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Rhinovirus (RV), the causative agent of the common cold, causes only mild upper respiratory tract infections in healthy individuals, but can cause longer lasting and more severe pulmonary infections in people with chronic lung diseases and in the setting of immune suppression or immune deficiency. RV-infected lung structural cells release type I interferon (IFN-I), initiating the immune response, leading to protection against viruses in conjunction with migratory immune cells. However, IFN-I release is deficient in some people with asthma. Innate immune cells, such as natural killer (NK) cells, are proposed to play major roles in the control of viral infections, and may contribute to exacerbations of chronic lung diseases, such as asthma. In this study, we characterized the NK cell response to RV infection using an in vitro model of infection in healthy individuals, and determined the extent to which IFN-I signaling mediates this response. The results indicate that RV stimulation in vitro induces NK cell activation in healthy donors, leading to degranulation and the release of cytotoxic mediators and cytokines. IFN-I signaling was partly responsible for NK cell activation and functional responses to RV. Overall, our findings suggest the involvement of NK cells in the control of RV infection in healthy individuals. Further understanding of NK cell regulation may deepen our understanding of the mechanisms that contribute to susceptibility to RV infections in asthma and other chronic lung diseases.
Collapse
Affiliation(s)
- Saskia L van der Heide
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Yang Xi
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - John W Upham
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
20
|
Cross RW, Prasad AN, Borisevich V, Geisbert JB, Agans KN, Deer DJ, Fenton KA, Geisbert TW. Crimean-Congo hemorrhagic fever virus strains Hoti and Afghanistan cause viremia and mild clinical disease in cynomolgus monkeys. PLoS Negl Trop Dis 2020; 14:e0008637. [PMID: 32790668 PMCID: PMC7447009 DOI: 10.1371/journal.pntd.0008637] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 08/25/2020] [Accepted: 07/24/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Development of vaccines and therapies against Crimean-Congo hemorrhagic fever virus (CCHFV) have been hindered by the lack of immunocompetent animal models. Recently, a lethal nonhuman primate model based on the CCHFV Hoti strain was reported. CCHFV Hoti caused severe disease in cynomolgus monkeys with 75% lethality when given by the intravenous (i.v.) route. METHODOLOGY/PRINCIPAL FINDINGS In a series of experiments, eleven cynomologus monkeys were exposed i.v. to CCHFV Hoti and four macaques were exposed i.v. to CCHFV Afghanistan. Despite transient viremia and changes in clinical pathology such as leukopenia and thrombocytopenia developing in all 15 animals, all macaques survived to the study endpoint without developing severe disease. CONCLUSIONS/SIGNIFICANCE We were unable to attribute differences in the results of our study versus the previous report to differences in the CCHFV Hoti stock, challenge dose, origin, or age of the macaques. The observed differences are most likely the result of the outbred nature of macaques and low animal numbers often used by necessity and for ethical considerations in BSL-4 studies. Nonetheless, while we were unable to achieve severe disease or lethality, the CCHFV Hoti and Afghanistan macaque models are useful for screening medical countermeasures using biomarkers including viremia and clinical pathology to assess efficacy.
Collapse
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Abhishek N. Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Joan B. Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Krystle N. Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Daniel J. Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Karla A. Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas W. Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
21
|
Ganjian H, Rajput C, Elzoheiry M, Sajjan U. Rhinovirus and Innate Immune Function of Airway Epithelium. Front Cell Infect Microbiol 2020; 10:277. [PMID: 32637363 PMCID: PMC7316886 DOI: 10.3389/fcimb.2020.00277] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Airway epithelial cells, which lines the respiratory mucosa is in direct contact with the environment. Airway epithelial cells are the primary target for rhinovirus and other inhaled pathogens. In response to rhinovirus infection, airway epithelial cells mount both pro-inflammatory responses and antiviral innate immune responses to clear the virus efficiently. Some of the antiviral responses include the expression of IFNs, endoplasmic reticulum stress induced unfolded protein response and autophagy. Airway epithelial cells also recruits other innate immune cells to establish antiviral state and resolve the inflammation in the lungs. In patients with chronic lung disease, these responses may be either defective or induced in excess leading to deficient clearing of virus and sustained inflammation. In this review, we will discuss the mechanisms underlying antiviral innate immunity and the dysregulation of some of these mechanisms in patients with chronic lung diseases.
Collapse
Affiliation(s)
- Haleh Ganjian
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| | - Charu Rajput
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| | - Manal Elzoheiry
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| | - Umadevi Sajjan
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
- Department of Physiology, Lewis Katz Medical School, Temple University, Philadelphia, PA, United States
| |
Collapse
|
22
|
Sauter A, Yi DH, Li Y, Roersma S, Appel S. The Culture Dish Surface Influences the Phenotype and Cytokine Production of Human Monocyte-Derived Dendritic Cells. Front Immunol 2019; 10:2352. [PMID: 31632415 PMCID: PMC6783514 DOI: 10.3389/fimmu.2019.02352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Monocyte-derived dendritic cells (moDC) are an important scientific and clinical source of functional dendritic cells (DC). However, the optimization of the generation process has to date mainly been limited to the variation of soluble factors. In this study, we investigated the impact of the cell culture dish surface on phenotype and cytokine profile. We compared a standard cell culture dish to a non-adherent culture dish for two immunogenic maturation conditions, two tolerogenic conditions, and an unstimulated control. Phenotype, cytokine profile and T cell stimulatory capacity were determined after a 3-day culture. Light microscopy revealed an increase in homotypic cluster formation correlated with the use of non-adherent surfaces, which could be reduced by using blocking antibodies against CD18. All surface markers analyzed showed moderate to strong differences depending on the culture dish surface, including significantly decreased expression of key maturation markers such as CD80, CD86, and CCR7 as well as PD-L1 on cells stimulated with the Jonuleit cytokine cocktail cultured on a non-adherent surface. Significant differences in the secretion of many cytokines were observed, especially for cells stimulated with LPS, with over 10-fold decreased secretion of IL-10, IL12-p40, and TNF-α from the cells cultured on the non-adherent surface. All immunogenic moDC populations showed similar capacity to induce antigen-specific T cells. These results provide evidence that the DC phenotype depends on the surface used during moDC generation. This has important implications for the optimization of DC-based immunotherapy development and underlines that the local surrounding can interfere with the final DC population beyond the soluble factors.
Collapse
Affiliation(s)
| | - Dag Heiro Yi
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Yayan Li
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Sabine Roersma
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Silke Appel
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
23
|
Unique transcriptional and protein-expression signature in human lung tissue-resident NK cells. Nat Commun 2019; 10:3841. [PMID: 31451696 PMCID: PMC6710242 DOI: 10.1038/s41467-019-11632-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
Human lung tissue-resident NK cells (trNK cells) are likely to play an important role in host responses towards viral infections, inflammatory conditions and cancer. However, detailed insights into these cells are still largely lacking. Here we show, using RNA sequencing and flow cytometry-based analyses, that subsets of human lung CD69+CD16− NK cells display hallmarks of tissue-residency, including high expression of CD49a, CD103, and ZNF683, and reduced expression of SELL, S1PR5, and KLF2/3. CD49a+CD16− NK cells are functionally competent, and produce IFN-γ, TNF, MIP-1β, and GM-CSF. After stimulation with IL-15, they upregulate perforin, granzyme B, and Ki67 to a similar degree as CD49a−CD16− NK cells. Comparing datasets from trNK cells in human lung and bone marrow with tissue-resident memory CD8+ T cells identifies core genes co-regulated either by tissue-residency, cell-type or location. Together, our data indicate that human lung trNK cells have distinct features, likely regulating their function in barrier immunity. Detailed characterizations of human lung tissue-resident natural killer (trNK) cells, which potentially regulate local immune responses, is still lacking. Here the authors show that lung CD69+ CD16– NK cells express tissue-residency markers, produce effector cytokines, and are distinct, feature-wise, from lung CD8+ memory T cells or trNK in other tissues.
Collapse
|
24
|
Panda SK, Colonna M. Innate Lymphoid Cells in Mucosal Immunity. Front Immunol 2019; 10:861. [PMID: 31134050 PMCID: PMC6515929 DOI: 10.3389/fimmu.2019.00861] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022] Open
Abstract
Innate lymphoid cells (ILCs) are innate counterparts of T cells that contribute to immune responses by secreting effector cytokines and regulating the functions of other innate and adaptive immune cells. ILCs carry out some unique functions but share some tasks with T cells. ILCs are present in lymphoid and non-lymphoid organs and are particularly abundant at the mucosal barriers, where they are exposed to allergens, commensal microbes, and pathogens. The impact of ILCs in mucosal immune responses has been extensively investigated in the gastrointestinal and respiratory tracts, as well as in the oral cavity. Here we review the state-of-the-art knowledge of ILC functions in infections, allergy and autoimmune disorders of the mucosal barriers.
Collapse
Affiliation(s)
- Santosh K Panda
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
25
|
Girkin J, Maltby S, Singanayagam A, Bartlett N, Mallia P. In vivo experimental models of infection and disease. RHINOVIRUS INFECTIONS 2019. [PMCID: PMC7149593 DOI: 10.1016/b978-0-12-816417-4.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human and animal models continue to play a crucial role in research to understand host immunity to rhinovirus (RV) and identify disease mechanisms. Human models have provided direct evidence that RV infection is capable of exacerbating chronic respiratory diseases and identified immunological processes that correlate with clinical disease outcomes. Mice are the most commonly used nonhuman experimental RV infection model. Although semipermissive, under defined experimental conditions sufficient replication occurs to induce host immune responses that recapitulate immunity and disease during human infection. The capacity to use genetically modified mouse strains and drug interventions has shown the mouse model to be an invaluable research tool defining causal relationships between host immunity and disease and supporting development of new treatments. Used in combination the insights achieved from human and animal experimental infection models provide complementary insights into RV biology and yield novel therapeutic options to reduce the burden of RV-induced disease.
Collapse
|
26
|
In the absence of natural killer cell activation donor-specific antibody mediates chronic, but not acute, kidney allograft rejection. Kidney Int 2018; 95:350-362. [PMID: 30503624 DOI: 10.1016/j.kint.2018.08.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 08/02/2018] [Accepted: 08/23/2018] [Indexed: 11/20/2022]
Abstract
Antibody mediated rejection (ABMR) is a major barrier to long-term kidney graft survival. Dysregulated donor-specific antibody (DSA) responses are induced in CCR5-deficient mice transplanted with complete major histocompatibility complex (MHC)-mismatched kidney allografts, and natural killer (NK) cells play a critical role in graft injury and rejection. We investigated the consequence of high DSA titers on kidney graft outcomes in the presence or absence of NK cell activation within the graft. Equivalent serum DSA titers were induced in CCR5-deficient B6 recipients of complete MHC mismatched A/J allografts and semi-allogeneic (A/J x B6) F1 kidney grafts, peaking by day 14 post-transplant. A/J allografts were rejected between days 16-28, whereas B6 isografts and semi-allogeneic grafts survived past day 65. On day 7 post-transplant, NK cell infiltration into A/J allografts was composed of distinct populations expressing high and low levels of the surface antigen NK1.1, with NK1.1low cells reflecting the highest level of activation. These NK cell populations increased with time post-transplant. In contrast, NK cell infiltration into semi-allogeneic grafts on day 7 was composed entirely of NK1.1high cells that decreased thereafter. On day 65 post-transplant the semi-allogeneic grafts had severe interstitial fibrosis, glomerulopathy, and arteriopathy, accompanied by expression of pro-fibrogenic genes. These results suggest that NK cells synergize with DSA to cause acute kidney allograft rejection, whereas high DSA titers in the absence of NK cell activation cannot provoke acute ABMR but instead induce the indolent development of interstitial fibrosis and glomerular injury that leads to late graft failure.
Collapse
|
27
|
Ling KM, Garratt LW, Lassmann T, Stick SM, Kicic A. Elucidating the Interaction of CF Airway Epithelial Cells and Rhinovirus: Using the Host-Pathogen Relationship to Identify Future Therapeutic Strategies. Front Pharmacol 2018; 9:1270. [PMID: 30464745 PMCID: PMC6234657 DOI: 10.3389/fphar.2018.01270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/17/2018] [Indexed: 01/07/2023] Open
Abstract
Chronic lung disease remains the primary cause of mortality in cystic fibrosis (CF). Growing evidence suggests respiratory viral infections are often more severe in CF compared to healthy peers and contributes to pulmonary exacerbations (PEx) and deterioration of lung function. Rhinovirus is the most prevalent respiratory virus detected, particularly during exacerbations in children with CF <5 years old. However, even though rhinoviral infections are likely to be one of the factors initiating the onset of CF lung disease, there is no effective targeted treatment. A better understanding of the innate immune responses by CF airway epithelial cells, the primary site of infection for viruses, is needed to identify why viral infections are more severe in CF. The aim of this review is to present the clinical impact of virus infection in both young children and adults with CF, focusing on rhinovirus infection. Previous in vitro and in vivo investigations looking at the mechanisms behind virus infection will also be summarized. The review will finish on the potential of transcriptomics to elucidate the host-pathogen responses by CF airway cells to viral infection and identify novel therapeutic targets.
Collapse
Affiliation(s)
- Kak-Ming Ling
- Paediatrics, Medical School, Faculty of Healthy and Medical Science, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Luke W Garratt
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Timo Lassmann
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Stephen M Stick
- Paediatrics, Medical School, Faculty of Healthy and Medical Science, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, WA, Australia
| | - Anthony Kicic
- Paediatrics, Medical School, Faculty of Healthy and Medical Science, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, WA, Australia
| | | | | | | |
Collapse
|
28
|
Dong X, Zhong N, Fang Y, Cai Q, Lu M, Lu Q. MicroRNA 27b-3p Modulates SYK in Pediatric Asthma Induced by Dust Mites. Front Pediatr 2018; 6:301. [PMID: 30406061 PMCID: PMC6204538 DOI: 10.3389/fped.2018.00301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 09/25/2018] [Indexed: 01/09/2023] Open
Abstract
The PI3K-AKT pathway is known to regulate cytokines in dust mite-induced pediatric asthma. However, the underlying molecular steps involved are not clear. In order to clarify further the molecular steps, this study investigated the expression of certain genes and the involvement of miRNAs in the PI3K-AKT pathway, which might affect the resultant cytokine-secretion. in-vivo and in-vitro ELISA, qRT-PCR and microarrays analyses were used in this study. A down-expression of miRNA-27b-3p in dust mite induced asthma group (group D) was found by microarray analysis. This was confirmed by qRT-PCR that found the miRNA-27b-3p transcripts that regulated the expression of SYK and EGFR were also significantly decreased (p < 0.01) in group D. The transcript levels of the SYK and PI3K genes were higher, while those of EGFR were lower in the former group. Meanwhile, we found significant differences in plasma concentrations of some cytokines between the dust mite-induced asthma subjects and the healthy controls. On the other hand, this correlated with the finding that the transcripts of SYK and its downstream PI3K were decreased in HBE transfected with miRNA-27b-3p, but were increased in HBE transfected with the inhibitor in vitro. Our results indicate that the differential expression of the miRNAs in dust mite-induced pediatric asthma may regulate their target gene SYK and may have an impact on the PI3K-AKT pathway associated with the production of cytokines. These findings should add new insight into the pathogenesis of pediatric asthma.
Collapse
Affiliation(s)
- Xiaoyan Dong
- Department of Pulmonary, Shanghai Children's Hospital, Shanghai, China
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai, China
- Shanghai Children's Hospital, Shanghai, China
| | - Nanbert Zhong
- Shanghai Children's Hospital, Shanghai, China
- Department of Human Genetics, Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
- Chinese Alliance of Translational Medicine for Maternal and Children's Health, Beijing, China
- Peking University Center of Medical Genetics, Peking University Health Science Centre, Beijing, China
| | - Yudan Fang
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai, China
- Shanghai Children's Hospital, Shanghai, China
| | - Qin Cai
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai, China
- Shanghai Children's Hospital, Shanghai, China
| | - Min Lu
- Department of Pulmonary, Shanghai Children's Hospital, Shanghai, China
- Shanghai Children's Hospital, Shanghai, China
| | - Quan Lu
- Department of Pulmonary, Shanghai Children's Hospital, Shanghai, China
- Shanghai Children's Hospital, Shanghai, China
| |
Collapse
|
29
|
Turi KN, Shankar J, Anderson LJ, Rajan D, Gaston K, Gebretsadik T, Das SR, Stone C, Larkin EK, Rosas-Salazar C, Brunwasser SM, Moore ML, Peebles RS, Hartert TV. Infant Viral Respiratory Infection Nasal Immune-Response Patterns and Their Association with Subsequent Childhood Recurrent Wheeze. Am J Respir Crit Care Med 2018; 198:1064-1073. [PMID: 29733679 PMCID: PMC6221572 DOI: 10.1164/rccm.201711-2348oc] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/07/2018] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Recurrent wheeze and asthma are thought to result from alterations in early life immune development following respiratory syncytial virus (RSV) infection. However, prior studies of the nasal immune response to infection have assessed only individual cytokines, which does not capture the whole spectrum of response to infection. OBJECTIVES To identify nasal immune phenotypes in response to RSV infection and their association with recurrent wheeze. METHODS A birth cohort of term healthy infants born June to December were recruited and followed to capture the first infant RSV infection. Nasal wash samples were collected during acute respiratory infection, viruses were identified by RT-PCR, and immune-response analytes were assayed using a multianalyte bead-based panel. Immune-response clusters were identified using machine learning, and association with recurrent wheeze at age 1 and 2 years was assessed using logistic regression. MEASUREMENTS AND MAIN RESULTS We identified two novel and distinct immune-response clusters to RSV and human rhinovirus. In RSV-infected infants, a nasal immune-response cluster characterized by lower non-IFN antiviral immune-response mediators, and higher type-2 and type-17 cytokines was significantly associated with first and second year recurrent wheeze. In comparison, we did not observe this in infants with human rhinovirus acute respiratory infection. Based on network analysis, type-2 and type-17 cytokines were central to the immune response to RSV, whereas growth factors and chemokines were central to the immune response to human rhinovirus. CONCLUSIONS Distinct immune-response clusters during infant RSV infection and their association with risk of recurrent wheeze provide insights into the risk factors for and mechanisms of asthma development.
Collapse
Affiliation(s)
- Kedir N. Turi
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Jyoti Shankar
- Infectious Disease Group, J. Craig Venter Institute, Rockville, Maryland; and
| | | | - Devi Rajan
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Kelsey Gaston
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | | | - Suman R. Das
- Division of Infectious Diseases, Department of Medicine
- Infectious Disease Group, J. Craig Venter Institute, Rockville, Maryland; and
| | - Cosby Stone
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Emma K. Larkin
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | | | | | - Martin L. Moore
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | | | - Tina V. Hartert
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| |
Collapse
|
30
|
To KKW, Lu L, Fong CHY, Wu AKL, Mok KY, Yip CCY, Ke YH, Sze KH, Lau SKP, Hung IFN, Yuen KY. Rhinovirus respiratory tract infection in hospitalized adult patients is associated with T H2 response irrespective of asthma. J Infect 2018; 76:465-474. [PMID: 29454786 DOI: 10.1016/j.jinf.2018.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/06/2018] [Accepted: 02/09/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES We assessed the immunological response of hospitalized adult patients with rhinovirus infection, including critically-ill patients. METHODS The differential white blood cell (WBC) count and the levels of 29 plasma cytokines/chemokines were compared between 50 adult hospitalized patients with rhinovirus infection and 100 age-matched controls with influenza virus infection. RESULTS The demographics and comorbidities were similar between rhinovirus and influenza patients, but severe disease was more common for the rhinovirus cohort. Rhinovirus patients had significantly higher WBC counts than influenza patients, especially for eosinophil (P = 3.1 × 10-8). The level of the TH2 cytokine IL-5 was significantly higher among rhinovirus patients, while the levels of 9 other cytokines/chemokines were significantly lower among rhinovirus patients. The levels of CXCL-10 (IP-10), CCL-2 (MCP-1), IFN-α2, IFN-γ, IL-10, and IL-15 remained significantly lower among rhinovirus patients after correction for multiple comparisons. Notably, CXCL-10 had the highest area under the receiver operating characteristic curve (AUC) in differentiating rhinovirus from influenza patients (AUC, 0.918). In the patient subgroup without asthma, the difference in the WBC count and cytokine/chemokine levels between rhinovirus and influenza patients remained statistically significant. CONCLUSIONS Rhinovirus infection was characterized by a prominent TH2 response, even in patients without asthma. CXCL-10 (IP-10) is a potential biomarker in differentiating rhinovirus from influenza infection.
Collapse
Affiliation(s)
- Kelvin K W To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Queen Mary Hospital, Hong Kong, China
| | - Lu Lu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carol H Y Fong
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Alan K L Wu
- Department of Pathology, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| | - Ka-Yi Mok
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cyril C Y Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Queen Mary Hospital, Hong Kong, China
| | - Yi-Hong Ke
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kong-Hung Sze
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China
| | - Susanna K P Lau
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Queen Mary Hospital, Hong Kong, China
| | - Ivan F N Hung
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China; Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Queen Mary Hospital, Hong Kong, China.
| |
Collapse
|
31
|
The Interplay between Natural Killer Cells and Human Herpesvirus-6. Viruses 2017; 9:v9120367. [PMID: 29194419 PMCID: PMC5744142 DOI: 10.3390/v9120367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Human Herpesvirus 6 (HHV-6) is a set of two closely related herpes viruses known as HHV-6A and HHV-6B. Both are lymphotropic viruses that establish latency in the host. The ability to evade the immune responses of effector cells is likely a major factor contributing to the development of a persistent HHV-6A/B (collectively termed HHV-6) infection. Natural killer (NK) cells are lymphocytes that, along with neutrophils and monocytes/macrophages, participate in the critical innate immune response during viral infections, but can also mediate the antigen-specific memory responses generally associated with adaptive immunity. NK cells compose the first barrier that viruses must break through to continue replication and dissemination, and a weak NK cell response may predispose an individual to chronic viral infections. Both HHV-6A and HHV-6B can interfere with NK cell-mediated anti-viral responses but the mechanisms by which each of these viruses affect NK cell activity differs. In this review, we will explore the nuanced relationships between the two viruses and NK cells, discussing, in addition, relevant disease associations.
Collapse
|
32
|
Xi Y, Troy NM, Anderson D, Pena OM, Lynch JP, Phipps S, Bosco A, Upham JW. Critical Role of Plasmacytoid Dendritic Cells in Regulating Gene Expression and Innate Immune Responses to Human Rhinovirus-16. Front Immunol 2017; 8:1351. [PMID: 29118754 PMCID: PMC5660993 DOI: 10.3389/fimmu.2017.01351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022] Open
Abstract
Though human rhinoviruses (HRVs) are usually innocuous viruses, they can trigger serious consequences in certain individuals, especially in the setting of impaired interferon (IFN) synthesis. Plasmacytoid dendritic cells (pDCs) are key IFN producing cells, though we know little about the role of pDC in HRV-induced immune responses. Herein, we used gene expression microarrays to examine HRV-activated peripheral blood mononuclear cells (PBMCs) from healthy people, in combination with pDC depletion, to assess whether observed gene expression patterns were pDC dependent. As expected, pDC depletion led to a major reduction in IFN-α release. This was associated with profound differences in gene expression between intact PBMC and pDC-depleted PBMC, and major changes in upstream regulators: 70–80% of the HRV activated genes appeared to be pDC dependent. Real-time PCR confirmed key changes in gene expression, in which the following selected genes were shown to be highly pDC dependent: the transcription factor IRF7, both IL-27 chains (IL-27p28 and EBI3), the alpha chain of the IL-15 receptor (IL-15RA) and the IFN-related gene IFI27. HRV-induced IL-6, IFN-γ, and IL-27 protein synthesis were also highly pDC dependent. Supplementing pDC-depleted cultures with recombinant IL-15, IFN-γ, IL-27, or IL-6 was able to restore the IFN-α response, thereby compensating for the absence of pDC. Though pDC comprise only a minority population of migratory leukocytes, our findings highlight the profound extent to which these cells contribute to the immune response to HRV.
Collapse
Affiliation(s)
- Yang Xi
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Niamh M Troy
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Denise Anderson
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Olga M Pena
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jason P Lynch
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Simon Phipps
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Anthony Bosco
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - John W Upham
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
33
|
Głobińska A, Kowalski ML. Innate lymphoid cells: the role in respiratory infections and lung tissue damage. Expert Rev Clin Immunol 2017; 13:991-999. [DOI: 10.1080/1744666x.2017.1366314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Anna Głobińska
- Department of Immunology, Rheumatology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Marek L Kowalski
- Department of Immunology, Rheumatology and Allergy, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
34
|
Hansel TT, Tunstall T, Trujillo-Torralbo MB, Shamji B, Del-Rosario A, Dhariwal J, Kirk PDW, Stumpf MPH, Koopmann J, Telcian A, Aniscenko J, Gogsadze L, Bakhsoliani E, Stanciu L, Bartlett N, Edwards M, Walton R, Mallia P, Hunt TM, Hunt TL, Hunt DG, Westwick J, Edwards M, Kon OM, Jackson DJ, Johnston SL. A Comprehensive Evaluation of Nasal and Bronchial Cytokines and Chemokines Following Experimental Rhinovirus Infection in Allergic Asthma: Increased Interferons (IFN-γ and IFN-λ) and Type 2 Inflammation (IL-5 and IL-13). EBioMedicine 2017; 19:128-138. [PMID: 28373098 PMCID: PMC5440599 DOI: 10.1016/j.ebiom.2017.03.033] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/09/2017] [Accepted: 03/24/2017] [Indexed: 01/04/2023] Open
Abstract
Background Rhinovirus infection is a major cause of asthma exacerbations. Objectives We studied nasal and bronchial mucosal inflammatory responses during experimental rhinovirus-induced asthma exacerbations. Methods We used nasosorption on days 0, 2–5 and 7 and bronchosorption at baseline and day 4 to sample mucosal lining fluid to investigate airway mucosal responses to rhinovirus infection in patients with allergic asthma (n = 28) and healthy non-atopic controls (n = 11), by using a synthetic absorptive matrix and measuring levels of 34 cytokines and chemokines using a sensitive multiplex assay. Results Following rhinovirus infection asthmatics developed more upper and lower respiratory symptoms and lower peak expiratory flows compared to controls (all P < 0.05). Asthmatics also developed higher nasal lining fluid levels of an anti-viral pathway (including IFN-γ, IFN-λ/IL-29, CXCL11/ITAC, CXCL10/IP10 and IL-15) and a type 2 inflammatory pathway (IL-4, IL-5, IL-13, CCL17/TARC, CCL11/eotaxin, CCL26/eotaxin-3) (area under curve day 0–7, all P < 0.05). Nasal IL-5 and IL-13 were higher in asthmatics at day 0 (P < 0.01) and levels increased by days 3 and 4 (P < 0.01). A hierarchical correlation matrix of 24 nasal lining fluid cytokine and chemokine levels over 7 days demonstrated expression of distinct interferon-related and type 2 pathways in asthmatics. In asthmatics IFN-γ, CXCL10/IP10, CXCL11/ITAC, IL-15 and IL-5 increased in bronchial lining fluid following viral infection (all P < 0.05). Conclusions Precision sampling of mucosal lining fluid identifies robust interferon and type 2 responses in the upper and lower airways of asthmatics during an asthma exacerbation. Nasosorption and bronchosorption have potential to define asthma endotypes in stable disease and at exacerbation. Following rhinovirus infection asthmatics have increased interferons and type 2 inflammation in airway mucosal lining fluid. Nasosorption cytokines and chemokines showed distinct pathways of interferon and type 2 inflammation in asthma. Precision mucosal sampling has potential for stratifying molecular endotypes of asthma. Validation of nasosorption and bronchosorption will be required for selection of asthmatics for therapy with biologics.
Experimental human rhinovirus (HRV) infection causes more severe upper and lower respiratory tract symptoms in allergic asthmatics than in healthy controls. There is greater induction of cytokines and chemokines in nasal and bronchial mucosal lining fluid (MLF) of asthmatics: with distinct pathways of type 2 and anti-viral/regulatory inflammation. Subject to further validation, analysis of MLF may prove useful in stratification of patients with asthma, and the definition of molecular endotypes. Interpretation Nasosorption and bronchosorption are precision sampling methods with potential for widespread application in respiratory and other mucosal diseases (e.g. gastrointestinal diseases). Biomarkers identified in nasosorption and bronchosorption samples will need to be validated compared to established airway sampling methods, in a range of asthma phenotypes, and with current and novel therapies.
Collapse
Affiliation(s)
- Trevor T Hansel
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK.
| | - Tanushree Tunstall
- Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK
| | - Maria-Belen Trujillo-Torralbo
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Betty Shamji
- Novartis Institute for Biomedical Research, Horsham, UK
| | - Ajerico Del-Rosario
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Jaideep Dhariwal
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | | | - Jens Koopmann
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; Medimmune, Cambridge, UK
| | - Aurica Telcian
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Leila Gogsadze
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Eteri Bakhsoliani
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Luminita Stanciu
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Nathan Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Michael Edwards
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Ross Walton
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Patrick Mallia
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Toby M Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - Trevor L Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - Duncan G Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - John Westwick
- Novartis Institute for Biomedical Research, Horsham, UK
| | | | - Onn Min Kon
- Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK
| | - David J Jackson
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Guy's and St Thomas' NHS Trust
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| |
Collapse
|
35
|
Pellom ST, Dudimah DF, Thounaojam MC, Uzhachenko RV, Singhal A, Richmond A, Shanker A. Bortezomib augments lymphocyte stimulatory cytokine signaling in the tumor microenvironment to sustain CD8+T cell antitumor function. Oncotarget 2017; 8:8604-8621. [PMID: 28052005 PMCID: PMC5352426 DOI: 10.18632/oncotarget.14365] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/07/2016] [Indexed: 12/19/2022] Open
Abstract
Tumor-induced immune tolerance poses a major challenge for therapeutic interventions aimed to manage cancer. We explored approaches to overcome T-cell suppression in murine breast and kidney adenocarcinomas, and lung fibrosarcoma expressing immunogenic antigens. We observed that treatment with a reversible proteasome inhibitor bortezomib (1 mg/kg body weight) in tumor-bearing mice significantly enhanced the expression of lymphocyte-stimulatory cytokines IL-2, IL-12, and IL-15. Notably, bortezomib administration reduced pulmonary nodules of mammary adenocarcinoma 4T1.2 expressing hemagglutinin (HA) model antigen (4T1HA) in mice. Neutralization of IL-12 and IL-15 cytokines with a regimen of blocking antibodies pre- and post-adoptive transfer of low-avidity HA518-526-specific CD8+T-cells following intravenous injection of 4T1HA cells increased the number of pulmonary tumor nodules. This neutralization effect was counteracted by the tumor metastasis-suppressing action of bortezomib treatments. In bortezomib-treated 4T1HA tumor-bearing mice, CD4+T-cells showed increased IL-2 production, CD11c+ dendritic cells showed increased IL-12 and IL-15 production, and HA-specific activated CD8+T-cells showed enhanced expression of IFNγ, granzyme-B and transcription factor eomesodermin. We also noted a trend of increased expression of IL-2, IL-12 and IL-15 receptors as well as increased phosphorylation of STAT5 in tumor-infiltrating CD8+T-cells following bortezomib treatment. Furthermore, bortezomib-treated CD8+T-cells showed increased phosphorylation of mitogen-activated protein kinase p38, and Akt, which was abrogated by phosphatidylinositide 3-kinase (PI3K) inhibitor. These data support the therapeutic potential of bortezomib in conjunction with other immunotherapies to augment the strength of convergent signals from CD8+T-cell signaling molecules including TCR, cytokine receptors and downstream PI3K/Akt/STAT5 pathways to sustain CD8+T-cell effector function in the tumor microenvironment.
Collapse
Affiliation(s)
- Samuel T. Pellom
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA
| | - Duafalia F. Dudimah
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Menaka C. Thounaojam
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Roman V. Uzhachenko
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Ashutosh Singhal
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Veterans Affairs, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Translational and Clinical Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Center for Translational and Clinical Immunology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
36
|
Glanville N, Peel TJ, Schröder A, Aniscenko J, Walton RP, Finotto S, Johnston SL. Tbet Deficiency Causes T Helper Cell Dependent Airways Eosinophilia and Mucus Hypersecretion in Response to Rhinovirus Infection. PLoS Pathog 2016; 12:e1005913. [PMID: 27683080 PMCID: PMC5040449 DOI: 10.1371/journal.ppat.1005913] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/04/2016] [Indexed: 11/18/2022] Open
Abstract
Current understanding of adaptive immune, particularly T cell, responses to human rhinoviruses (RV) is limited. Memory T cells are thought to be of a primarily T helper 1 type, but both T helper 1 and T helper 2 memory cells have been described, and heightened T helper 2/ lessened T helper 1 responses have been associated with increased RV-induced asthma exacerbation severity. We examined the contribution of T helper 1 cells to RV-induced airways inflammation using mice deficient in the transcription factor T-Box Expressed In T Cells (Tbet), a critical controller of T helper 1 cell differentiation. Using flow cytometry we showed that Tbet deficient mice lacked the T helper 1 response of wild type mice and instead developed mixed T helper 2/T helper 17 responses to RV infection, evidenced by increased numbers of GATA binding protein 3 (GATA-3) and RAR-related orphan receptor gamma t (RORγt), and interleukin-13 and interleukin-17A expressing CD4+ T cells in the lung. Forkhead box P3 (FOXP3) and interleukin-10 expressing T cell numbers were unaffected. Tbet deficient mice also displayed deficiencies in lung Natural Killer, Natural Killer T cell and γδT cell responses, and serum neutralising antibody responses. Tbet deficient mice exhibited pronounced airways eosinophilia and mucus production in response to RV infection that, by utilising a CD4+ cell depleting antibody, were found to be T helper cell dependent. RV induction of T helper 2 and T helper 17 responses may therefore have an important role in directly driving features of allergic airways disease such as eosinophilia and mucus hypersecretion during asthma exacerbations.
Collapse
Affiliation(s)
- Nicholas Glanville
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Tamlyn J. Peel
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Armin Schröder
- Laboratory of Cellular and Molecular Lung Immunology, Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Ross P. Walton
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Susetta Finotto
- Laboratory of Cellular and Molecular Lung Immunology, Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian L. Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
Complementary Effects of Interleukin-15 and Alpha Interferon Induce Immunity in Hepatitis B Virus Transgenic Mice. J Virol 2016; 90:8563-74. [PMID: 27440883 PMCID: PMC5021417 DOI: 10.1128/jvi.01030-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/12/2016] [Indexed: 12/17/2022] Open
Abstract
In chronic hepatitis B (CHB), failure to control hepatitis B virus (HBV) is associated with T cell dysfunction. HBV transgenic mice mirror many features of the human disease, including T cell unresponsiveness, and thus represent an appropriate model in which to test novel therapeutic strategies. To date, the tolerant state of CD8+ T cells in these animals could be altered only by strong immunogens or by immunization with HBV antigen-pulsed dendritic cells; however, the effectors induced were unable to suppress viral gene expression or replication. Because of the known stimulatory properties of alpha interferon (IFN-α) and interleukin-15 (IL-15), this study explored the therapeutic potential of liver-directed gene transfer of these cytokines in a murine model of CHB using adeno-associated virus (AAV) delivery. This combination not only resulted in a reduction in the viral load in the liver and the induction of an antibody response but also gave rise to functional and specific CD8+ immunity. Furthermore, when splenic and intrahepatic lymphocytes from IFN-α- and IL-15-treated animals were transferred to new HBV carriers, partial antiviral immunity was achieved. In contrast to previous observations made using either cytokine alone, markedly attenuated PD-L1 induction in hepatic tissue was observed upon coadministration. An initial study with CHB patient samples also gave promising results. Hence, we demonstrated synergy between two stimulating cytokines, IL-15 and IFN-α, which, given together, constitute a potent approach to significantly enhance the CD8+ T cell response in a state of immune hyporesponsiveness. Such an approach may be useful for treating chronic viral infections and neoplastic conditions. IMPORTANCE With 350 million people affected worldwide and 600,000 annual deaths due to HBV-induced liver cirrhosis and/or hepatocellular carcinoma, chronic hepatitis B (CHB) is a major health problem. However, current treatment options are costly and not very effective and/or need to be administered for life. The unprecedented efficacy of the strategy described in our paper may offer an alternative and is relevant for a broad spectrum of readers because of its clear translational importance to other chronic viral infections in which a hyporesponsive antigen-specific T cell repertoire prevents clearance of the pathogen.
Collapse
|
38
|
Abstract
Human rhinovirus (HRV) and coronavirus (HCoV) infections are associated with both upper respiratory tract illness (“the common cold”) and lower respiratory tract illness (pneumonia). New species of HRVs and HCoVs have been diagnosed in the past decade. More sensitive diagnostic tests such as reverse transcription-polymerase chain reaction have expanded our understanding of the role these viruses play in both immunocompetent and immunosuppressed hosts. Recent identification of severe acute respiratory syndrome and Middle East respiratory syndrome viruses causing serious respiratory illnesses has led to renewed efforts for vaccine development. The role these viruses play in patients with chronic lung disease such as asthma makes the search for antiviral agents of increased importance.
Collapse
Affiliation(s)
- Stephen B Greenberg
- Department of Medicine, Ben Taub Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
39
|
Affiliation(s)
- J. E. Gern
- Pediatrics and Medicine; University of Wisconsin-Madison; Madison WI USA
| |
Collapse
|
40
|
Holtzman MJ, Byers DE, Alexander-Brett J, Wang X. The role of airway epithelial cells and innate immune cells in chronic respiratory disease. Nat Rev Immunol 2014; 14:686-98. [PMID: 25234144 PMCID: PMC4782595 DOI: 10.1038/nri3739] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An abnormal immune response to environmental agents is generally thought to be responsible for causing chronic respiratory diseases, such as asthma and chronic obstructive pulmonary disease (COPD). Based on studies of experimental models and human subjects, there is increasing evidence that the response of the innate immune system is crucial for the development of this type of airway disease. Airway epithelial cells and innate immune cells represent key components of the pathogenesis of chronic airway disease and are emerging targets for new therapies. In this Review, we summarize the innate immune mechanisms by which airway epithelial cells and innate immune cells regulate the development of chronic respiratory diseases. We also explain how these pathways are being targeted in the clinic to treat patients with these diseases.
Collapse
Affiliation(s)
- Michael J Holtzman
- 1] Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA. [2] Department of Cell Biology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | - Jennifer Alexander-Brett
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | - Xinyu Wang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| |
Collapse
|
41
|
Woo Y, Jeong D, Chung DH, Kim HY. The roles of innate lymphoid cells in the development of asthma. Immune Netw 2014; 14:171-81. [PMID: 25177249 PMCID: PMC4148487 DOI: 10.4110/in.2014.14.4.171] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/24/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023] Open
Abstract
Asthma is a common pulmonary disease with several different forms. The most studied form of asthma is the allergic form, which is mainly related to the function of Th2 cells and their production of cytokines (IL-4, IL-5, and IL-13) in association with allergen sensitization and adaptive immunity. Recently, there have been many advances in understanding non-allergic asthma, which seems to be related to environmental factors such as air pollution, infection, or even obesity. Cells of the innate immune system, including macrophages, neutrophils, and natural killer T cells as well as the newly described innate lymphoid cells, are effective producers of a variety of cytokines and seem to play important roles in the development of non-allergic asthma. In this review, we focus on recent findings regarding innate lymphoid cells and their roles in asthma.
Collapse
Affiliation(s)
- Yeonduk Woo
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Dongjin Jeong
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Doo Hyun Chung
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Hye Young Kim
- Department of Medical Science, Seoul National University College of Medicine and Hospital, Seoul 110-744, Korea
| |
Collapse
|
42
|
NK cells in mucosal defense against infection. BIOMED RESEARCH INTERNATIONAL 2014; 2014:413982. [PMID: 25197644 PMCID: PMC4150440 DOI: 10.1155/2014/413982] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 07/31/2014] [Indexed: 01/06/2023]
Abstract
Conventional natural killer cells (NK cells) provide continual surveillance for cancer and rapid responses to infection. They develop in the bone marrow, emerge as either NK precursor cells, immature, or mature cells, and disperse throughout the body. In the periphery NK cells provide critical defense against pathogens and cancer and are noted to develop features of adaptive immune responses. In the tightly regulated and dynamic mucosal tissues, they set up residency via unknown mechanisms and from sources that are yet to be defined. Once resident, they appear to have the ability to functionally mature dependent on the mucosal tissue microenvironment. Mucosal NK cells play a pivotal role in early protection through their cytolytic function and IFNγ production against bacteria, fungi, viruses, and parasitic infections. This review presents what is known about NK cell development and phenotypes of mucosal tissue resident conventional NK cells. The question of how they come to reside in their tissues and published data on their function against pathogens during mucosal infection are discussed. Dissecting major questions highlighted in this review will be important to the further understanding of NK cell homing and functional diversity and improve rational design of NK cell based therapies against mucosal infection.
Collapse
|