51
|
Zhang X, Feng WH. Porcine Reproductive and Respiratory Syndrome Virus Evades Antiviral Innate Immunity via MicroRNAs Regulation. Front Microbiol 2022; 12:804264. [PMID: 34975824 PMCID: PMC8714953 DOI: 10.3389/fmicb.2021.804264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most important diseases in pigs, leading to significant economic losses in the swine industry worldwide. MicroRNAs (miRNAs) are small single-stranded non-coding RNAs involved in regulating gene expressions at the post-transcriptional levels. A variety of host miRNAs are dysregulated and exploited by PRRSV to escape host antiviral surveillance and help virus infection. In addition, PRRSV might encode miRNAs. In this review, we will summarize current progress on how PRRSV utilizes miRNAs for immune evasions. Increasing knowledge of the role of miRNAs in immune evasion will improve our understanding of PRRSV pathogenesis and help us develop new treatments for PRRSV-associated diseases.
Collapse
Affiliation(s)
- Xuan Zhang
- State Key Laboratory of Agrobiotechnology, Ministry of Agriculture Key Laboratory of Soil Microbiology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wen-Hai Feng
- State Key Laboratory of Agrobiotechnology, Ministry of Agriculture Key Laboratory of Soil Microbiology, Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
52
|
Talker SC, Barut GT, Lischer HE, Rufener R, von Münchow L, Bruggmann R, Summerfield A. Monocyte biology conserved across species: Functional insights from cattle. Front Immunol 2022; 13:889175. [PMID: 35967310 PMCID: PMC9373011 DOI: 10.3389/fimmu.2022.889175] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022] Open
Abstract
Similar to human monocytes, bovine monocytes can be split into CD14highCD16- classical, CD14highCD16high intermediate and CD14-/dimCD16high nonclassical monocytes (cM, intM, and ncM, respectively). Here, we present an in-depth analysis of their steady-state bulk- and single-cell transcriptomes, highlighting both pronounced functional specializations and transcriptomic relatedness. Bulk gene transcription indicates pro-inflammatory and antibacterial roles of cM, while ncM and intM appear to be specialized in regulatory/anti-inflammatory functions and tissue repair, as well as antiviral responses and T-cell immunomodulation. Notably, intM stood out by high expression of several genes associated with antigen presentation. Anti-inflammatory and antiviral functions of ncM are further supported by dominant oxidative phosphorylation and selective strong responses to TLR7/8 ligands, respectively. Moreover, single-cell RNA-seq revealed previously unappreciated heterogeneity within cM and proposes intM as a transient differentiation intermediate between cM and ncM.
Collapse
Affiliation(s)
- Stephanie C. Talker
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- *Correspondence: Stephanie C. Talker,
| | - G. Tuba Barut
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Heidi E.L. Lischer
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Reto Rufener
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
53
|
Du W, Frankel TL, Green M, Zou W. IFNγ signaling integrity in colorectal cancer immunity and immunotherapy. Cell Mol Immunol 2022; 19:23-32. [PMID: 34385592 PMCID: PMC8752802 DOI: 10.1038/s41423-021-00735-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of colorectal cancer patients are not responsive to immune checkpoint blockade (ICB). The interferon gamma (IFNγ) signaling pathway drives spontaneous and ICB-induced antitumor immunity. In this review, we summarize recent advances in the epigenetic, genetic, and functional integrity of the IFNγ signaling pathway in the colorectal cancer microenvironment and its immunological relevance in the therapeutic efficacy of and resistance to ICB. Moreover, we discuss how to target IFNγ signaling to inform novel clinical trials to treat patients with colorectal cancer.
Collapse
Affiliation(s)
- Wan Du
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Timothy L Frankel
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Michael Green
- Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Healthcare System, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Graduate Programs in Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Graduate Programs in Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Tumor Biology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
54
|
Korchagina AA, Koroleva E, Tumanov AV. Innate Lymphoid Cells in Response to Intracellular Pathogens: Protection Versus Immunopathology. Front Cell Infect Microbiol 2021; 11:775554. [PMID: 34938670 PMCID: PMC8685334 DOI: 10.3389/fcimb.2021.775554] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/03/2021] [Indexed: 12/23/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous group of cytokine-producing lymphocytes which are predominantly located at mucosal barrier surfaces, such as skin, lungs, and gastrointestinal tract. ILCs contribute to tissue homeostasis, regulate microbiota-derived signals, and protect against mucosal pathogens. ILCs are classified into five major groups by their developmental origin and distinct cytokine production. A recently emerged intriguing feature of ILCs is their ability to alter their phenotype and function in response to changing local environmental cues such as pathogen invasion. Once the pathogen crosses host barriers, ILCs quickly activate cytokine production to limit the spread of the pathogen. However, the dysregulated ILC responses can lead to tissue inflammation and damage. Furthermore, the interplay between ILCs and other immune cell types shapes the outcome of the immune response. Recent studies highlighted the important role of ILCs for host defense against intracellular pathogens. Here, we review recent advances in understanding the mechanisms controlling protective and pathogenic ILC responses to intracellular pathogens. This knowledge can help develop new ILC-targeted strategies to control infectious diseases and immunopathology.
Collapse
Affiliation(s)
- Anna A Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ekaterina Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
55
|
Czajka-Francuz P, Cisoń-Jurek S, Czajka A, Kozaczka M, Wojnar J, Chudek J, Francuz T. Systemic Interleukins' Profile in Early and Advanced Colorectal Cancer. Int J Mol Sci 2021; 23:124. [PMID: 35008550 PMCID: PMC8745135 DOI: 10.3390/ijms23010124] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 02/05/2023] Open
Abstract
Tumor microenvironment (TME) is characterized by mutual interactions of the tumor, stromal and immune cells. Early and advanced colorectal tumors differ in structure and present altered serum cytokine levels. Mutual crosstalk among TME infiltrating cells may shift the balance into immune suppressive or pro-inflammatory, antitumor response this way influencing patients' prognosis. Cancer-related inflammation affects all the body and this way, the systemic level of cytokines could reflect TME processes. Despite numerous studies, it is still not known how systemic cytokines levels change during colorectal cancer (CRC) tumor development. Better understanding tumor microenvironment processes could help in planning therapeutic interventions and more accurate patient prognosis. To contribute to the comprehension of these processes within TME, we reviewed cytokines levels from clinical trials in early and advanced colorectal cancer. Presented data were analyzed in the context of experimental studies and studies analyzing tumor infiltration with immune cells. The review summarizes clinical data of cytokines secreted by tumor microenvironment cells: lymphocytes T helper 1 (Th1), lymphocytes T helper 2 (Th2), lymphocytes T helper 17 (Th17), regulatory T cells (Treg cells), regulatory T cells (Breg cells), M1/M2 macrophages, N1/N2 neutrophils, myeloid-derived suppressor cells (MDSC), dendritic cells (DC), innate lymphoid cells (ILC) natural killer (NK) cells and tumor cells.
Collapse
Affiliation(s)
- Paulina Czajka-Francuz
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Sylwia Cisoń-Jurek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Aleksander Czajka
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-635 Katowice, Poland;
| | - Maciej Kozaczka
- Department of Radiotherapy and Chemotherapy, National Institute of Oncology, Public Research Institute in Gliwice, 44-101 Gliwice, Poland;
| | - Jerzy Wojnar
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Tomasz Francuz
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| |
Collapse
|
56
|
Wijers CDM, Pham L, Menon S, Boyd KL, Noel HR, Skaar EP, Gaddy JA, Palmer LD, Noto MJ. Identification of Two Variants of Acinetobacter baumannii Strain ATCC 17978 with Distinct Genotypes and Phenotypes. Infect Immun 2021; 89:e0045421. [PMID: 34460288 PMCID: PMC8594612 DOI: 10.1128/iai.00454-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 01/11/2023] Open
Abstract
Acinetobacter baumannii is a nosocomial pathogen that exhibits substantial genomic plasticity. Here, the identification of two variants of A. baumannii ATCC 17978 that differ based on the presence of a 44-kb accessory locus, named AbaAL44 (A. baumannii accessory locus 44 kb), is described. Analyses of existing deposited data suggest that both variants are found in published studies of A. baumannii ATCC 17978 and that American Type Culture Collection (ATCC)-derived laboratory stocks comprise a mix of these two variants. Yet, each variant exhibits distinct interactions with the host in vitro and in vivo. Infection with the variant that harbors AbaAL44 (A. baumannii 17978 UN) results in decreased bacterial burdens and increased neutrophilic lung inflammation in a mouse model of pneumonia, and affects the production of interleukin 1 beta (IL-1β) and IL-10 by infected macrophages. AbaAL44 harbors putative pathogenesis genes, including those predicted to encode a type I pilus cluster, a catalase, and a cardiolipin synthase. The accessory catalase increases A. baumannii resistance to oxidative stress and neutrophil-mediated killing in vitro. The accessory cardiolipin synthase plays a dichotomous role by promoting bacterial uptake and increasing IL-1β production by macrophages, but also by enhancing bacterial resistance to cell envelope stress. Collectively, these findings highlight the phenotypic consequences of the genomic dynamism of A. baumannii through the evolution of two variants of a common type strain with distinct infection-related attributes.
Collapse
Affiliation(s)
- Christiaan D. M. Wijers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ly Pham
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Swapna Menon
- AnalyzeDat Consulting Services, Ernakulam, Kerala, India
| | - Kelli L. Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hannah R. Noel
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jennifer A. Gaddy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee, USA
| | - Lauren D. Palmer
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Michael J. Noto
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
57
|
Targeting interferon-γ in hyperinflammation: opportunities and challenges. Nat Rev Rheumatol 2021; 17:678-691. [PMID: 34611329 DOI: 10.1038/s41584-021-00694-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Interferon-γ (IFNγ) is a pleiotropic cytokine with multiple effects on the inflammatory response and on innate and adaptive immunity. Overproduction of IFNγ underlies several, potentially fatal, hyperinflammatory or immune-mediated diseases. Several data from animal models and/or from translational research in patients point to a role of IFNγ in hyperinflammatory diseases, such as primary haemophagocytic lymphohistiocytosis, various forms of secondary haemophagocytic lymphohistiocytosis, including macrophage activation syndrome, and cytokine release syndrome, all of which are often managed by rheumatologists or in consultation with rheumatologists. Given the effects of IFNγ on B cells and T follicular helper cells, a role for IFNγ in systemic lupus erythematosus pathogenesis is emerging. To improve our understanding of the role of IFNγ in human disease, IFNγ-related biomarkers that are relevant for the management of hyperinflammatory diseases are progressively being identified and studied, especially because circulating levels of IFNγ do not always reflect its overproduction in tissue. These biomarkers include STAT1 (specifically the phosphorylated form), neopterin and the chemokine CXCL9. IFNγ-neutralizing agents have shown efficacy in the treatment of primary haemophagocytic lymphohistiocytosis in clinical trials and initial promising results have been obtained in various forms of secondary haemophagocytic lymphohistiocytosis, including macrophage activation syndrome. In clinical practice, there is a growing body of evidence supporting the usefulness of circulating CXCL9 levels as a biomarker reflecting IFNγ production.
Collapse
|
58
|
Yadav KK, Kenney SP. Hepatitis E Virus Immunopathogenesis. Pathogens 2021; 10:pathogens10091180. [PMID: 34578211 PMCID: PMC8465319 DOI: 10.3390/pathogens10091180] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 12/22/2022] Open
Abstract
Hepatitis E virus is an important emerging pathogen producing a lethal impact on the pregnant population and immunocompromised patients. Starting in 1983, it has been described as the cause for acute hepatitis transmitted via the fecal–oral route. However, zoonotic and blood transfusion transmission of HEV have been reported in the past few decades, leading to the detailed research of HEV pathogenesis. The reason behind HEV being highly virulent to the pregnant population particularly during the third trimester, leading to maternal and fetal death, remains unknown. Various host factors (immunological, nutritional, hormonal) and viral factors have been studied to define the key determinants assisting HEV to be virulent in pregnant and immunocompromised patients. Similarly, chronic hepatitis is seen particularly in solid organ transplant patients, resulting in fatal conditions. This review describes recent advances in the immunopathophysiology of HEV infections in general, pregnant, and immunocompromised populations, and further elucidates the in vitro and in vivo models utilized to understand HEV pathogenesis.
Collapse
|
59
|
Li D, Zhang J, Yang W, Li P, Ru Y, Kang W, Li L, Ran Y, Zheng H. African swine fever virus protein MGF-505-7R promotes virulence and pathogenesis by inhibiting JAK1- and JAK2-mediated signaling. J Biol Chem 2021; 297:101190. [PMID: 34517008 PMCID: PMC8526981 DOI: 10.1016/j.jbc.2021.101190] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 11/03/2022] Open
Abstract
African swine fever virus (ASFV) is a large DNA virus that is highly contagious and pathogenic in domestic pigs with a mortality rate up to 100%. However, how ASFV suppresses JAK-STAT1 signaling to evade the immune response remains unclear. In this study, we found that the ASFV-encoded protein MGF-505-7R inhibited proinflammatory IFN-γ-mediated JAK-STAT1 signaling. Mechanistically, MGF-505-7R was found to interact with JAK1 and JAK2 and mediate their degradation. Further study indicated that MGF-505-7R promoted degradation of JAK1 and JAK2 by upregulating the E3 ubiquitin ligase RNF125 expression and inhibiting expression of Hes5, respectively. Consistently, MGF-505-7R-deficient ASFV induced high levels of IRF1 expression and displayed compromised replication both in primary porcine alveolar macrophages and pigs compared with wild-type ASFV. Furthermore, MGF-505-7R deficiency attenuated the virulence of the ASFV and pathogenesis of ASF in pigs. These findings suggest that the JAK-STAT1 axis mediates the innate immune response to the ASFV and that MGF-505-7R plays a critical role in the virulence of the ASFV and pathogenesis of ASF by antagonizing this axis. Thus, we conclude that deletion of MGF-505-7R may serve as a strategy to develop attenuated vaccines against the ASFV.
Collapse
Affiliation(s)
- Dan Li
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jing Zhang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Wenping Yang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Pan Li
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yi Ru
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Weifang Kang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - LuLu Li
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yong Ran
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China.
| |
Collapse
|
60
|
Nouri-Goushki M, Isaakidou A, Eijkel BIM, Minneboo M, Liu Q, Boukany PE, Mirzaali MJ, Fratila-Apachitei LE, Zadpoor AA. 3D printed submicron patterns orchestrate the response of macrophages. NANOSCALE 2021; 13:14304-14315. [PMID: 34190291 PMCID: PMC8412028 DOI: 10.1039/d1nr01557e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/16/2021] [Indexed: 05/12/2023]
Abstract
The surface topography of engineered extracellular matrices is one of the most important physical cues regulating the phenotypic polarization of macrophages. However, not much is known about the ways through which submicron (i.e., 100-1000 nm) topographies modulate the polarization of macrophages. In the context of bone tissue regeneration, it is well established that this range of topographies stimulates the osteogenic differentiation of stem cells. Since the immune response affects the bone tissue regeneration process, the immunomodulatory consequences of submicron patterns should be studied prior to their clinical application. Here, we 3D printed submicron pillars (using two-photon polymerization technique) with different heights and interspacings to perform the first ever systematic study of such effects. Among the studied patterns, the highest degree of elongation was observed for the cells cultured on those with the tallest and densest pillars. After 3 days of culture with inflammatory stimuli (LPS/IFN-γ), sparsely decorated surfaces inhibited the expression of the pro-inflammatory cellular marker CCR7 as compared to day 1 and to the other patterns. Furthermore, sufficiently tall pillars polarized the M1 macrophages towards a pro-healing (M2) phenotype, as suggested by the expression of CD206 within the first 3 days. As some of the studied patterns are known to be osteogenic, the osteoimmunomodulatory capacity of the patterns should be further studied to optimize their bone tissue regeneration performance.
Collapse
Affiliation(s)
- M Nouri-Goushki
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| | - A Isaakidou
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| | - B I M Eijkel
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| | - M Minneboo
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| | - Q Liu
- Department of Chemical Engineering, Delft University of Technology (TU Delft), van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - P E Boukany
- Department of Chemical Engineering, Delft University of Technology (TU Delft), van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - M J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| | - L E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| | - A A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD, Delft, The Netherlands.
| |
Collapse
|
61
|
Houser MC, Caudle WM, Chang J, Kannarkat GT, Yang Y, Kelly SD, Oliver D, Joers V, Shannon KM, Keshavarzian A, Tansey MG. Experimental colitis promotes sustained, sex-dependent, T-cell-associated neuroinflammation and parkinsonian neuropathology. Acta Neuropathol Commun 2021; 9:139. [PMID: 34412704 PMCID: PMC8375080 DOI: 10.1186/s40478-021-01240-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background The etiology of sporadic Parkinson’s disease (PD) remains uncertain, but genetic, epidemiological, and physiological overlap between PD and inflammatory bowel disease suggests that gut inflammation could promote dysfunction of dopamine-producing neurons in the brain. Mechanisms behind this pathological gut-brain effect and their interactions with sex and with environmental factors are not well understood but may represent targets for therapeutic intervention. Methods We sought to identify active inflammatory mechanisms which could potentially contribute to neuroinflammation and neurological disease in colon biopsies and peripheral blood immune cells from PD patients. Then, in mouse models, we assessed whether dextran sodium sulfate-mediated colitis could exert lingering effects on dopaminergic pathways in the brain and whether colitis increased vulnerability to a subsequent exposure to the dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We assessed the involvement of inflammatory mechanisms identified in the PD patients in colitis-related neurological dysfunction in male and female mice, utilizing mice lacking the Regulator of G-Protein Signaling 10 (RGS10)—an inhibitor of nuclear factor kappa B (NFκB)—to model enhanced NFκB activity, and mice in which CD8+ T-cells were depleted. Results High levels of inflammatory markers including CD8B and NFκB p65 were found in colon biopsies from PD patients, and reduced levels of RGS10 were found in immune cells in the blood. Male mice that experienced colitis exhibited sustained reductions in tyrosine hydroxylase but not in dopamine as well as sustained CD8+ T-cell infiltration and elevated Ifng expression in the brain. CD8+ T-cell depletion prevented colitis-associated reductions in dopaminergic markers in males. In both sexes, colitis potentiated the effects of MPTP. RGS10 deficiency increased baseline intestinal inflammation, colitis severity, and neuropathology. Conclusions This study identifies peripheral inflammatory mechanisms in PD patients and explores their potential to impact central dopaminergic pathways in mice. Our findings implicate a sex-specific interaction between gastrointestinal inflammation and neurologic vulnerability that could contribute to PD pathogenesis, and they establish the importance of CD8+ T-cells in this process in male mice. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40478-021-01240-4.
Collapse
|
62
|
Blanco FC, Gravisaco MJ, Bigi MM, García EA, Marquez C, McNeil M, Jackson M, Bigi F. Identifying Bacterial and Host Factors Involved in the Interaction of Mycobacterium bovis with the Bovine Innate Immune Cells. Front Immunol 2021; 12:674643. [PMID: 34335572 PMCID: PMC8319915 DOI: 10.3389/fimmu.2021.674643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis is an important animal and zoonotic disease caused by Mycobacterium bovis. The innate immune response is the first line of defense against pathogens and is also crucial for the development of an efficient adaptive immune response. In this study we used an in vitro co-culture model of antigen presenting cells (APC) and autologous lymphocytes derived from peripheral blood mononuclear cells to identify the cell populations and immune mediators that participate in the development of an efficient innate response capable of controlling the intracellular replication of M. bovis. After M. bovis infection, bovine immune cell cultures displayed upregulated levels of iNOS, IL-22 and IFN-γ and the induction of the innate immune response was dependent on the presence of differentiated APC. Among the analyzed M. bovis isolates, only a live virulent M. bovis isolate induced an efficient innate immune response, which was increased upon stimulation of cell co-cultures with the M. bovis culture supernatant. Moreover, we demonstrated that an allelic variation of the early secreted protein ESAT-6 (ESAT6 T63A) expressed in the virulent strain is involved in this increased innate immune response. These results highlight the relevance of the compounds secreted by live M. bovis as well as the variability among the assessed M. bovis strains to induce an efficient innate immune response.
Collapse
Affiliation(s)
- Federico Carlos Blanco
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
- (Consejo Nacional de Investigaciones Científicas y Tecnológicas) National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - María José Gravisaco
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
| | - María Mercedes Bigi
- (Facultad de Agronomía, Universidad de Buenos Aires) School of Agronomy, University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Elizabeth Andrea García
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
| | - Cecilia Marquez
- High Technology Analytical Centre, Laboratory, Buenos Aires, Argentina
| | - Mike McNeil
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mary Jackson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Fabiana Bigi
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
- (Consejo Nacional de Investigaciones Científicas y Tecnológicas) National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
63
|
Budithi A, Su S, Kirshtein A, Shahriyari L. Data Driven Mathematical Model of FOLFIRI Treatment for Colon Cancer. Cancers (Basel) 2021; 13:2632. [PMID: 34071939 PMCID: PMC8198096 DOI: 10.3390/cancers13112632] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Many colon cancer patients show resistance to their treatments. Therefore, it is important to consider unique characteristic of each tumor to find the best treatment options for each patient. In this study, we develop a data driven mathematical model for interaction between the tumor microenvironment and FOLFIRI drug agents in colon cancer. Patients are divided into five distinct clusters based on their estimated immune cell fractions obtained from their primary tumors' gene expression data. We then analyze the effects of drugs on cancer cells and immune cells in each group, and we observe different responses to the FOLFIRI drugs between patients in different immune groups. For instance, patients in cluster 3 with the highest T-reg/T-helper ratio respond better to the FOLFIRI treatment, while patients in cluster 2 with the lowest T-reg/T-helper ratio resist the treatment. Moreover, we use ROC curve to validate the model using the tumor status of the patients at their follow up, and the model predicts well for the earlier follow up days.
Collapse
Affiliation(s)
- Aparajita Budithi
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (A.B.); (S.S.)
| | - Sumeyye Su
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (A.B.); (S.S.)
| | - Arkadz Kirshtein
- Department of Mathematics, Tufts University, Medford, MA 02155, USA;
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (A.B.); (S.S.)
| |
Collapse
|
64
|
The Effect of Inflammatory Priming on the Therapeutic Potential of Mesenchymal Stromal Cells for Spinal Cord Repair. Cells 2021; 10:cells10061316. [PMID: 34070547 PMCID: PMC8227154 DOI: 10.3390/cells10061316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSC) are used for cell therapy for spinal cord injury (SCI) because of their ability to support tissue repair by paracrine signaling. Preclinical and clinical research testing MSC transplants for SCI have revealed limited success, which warrants the exploration of strategies to improve their therapeutic efficacy. MSC are sensitive to the microenvironment and their secretome can be altered in vitro by exposure to different culture media. Priming MSC with inflammatory stimuli increases the expression and secretion of reparative molecules. We studied the effect of macrophage-derived inflammation priming on MSC transplants and of primed MSC (pMSC) acute transplants (3 days) on spinal cord repair using an adult rat model of moderate-severe contusive SCI. We found a decrease in long-term survival of pMSC transplants compared with unprimed MSC transplants. With a pMSC transplant, we found significantly more anti-inflammatory macrophages in the contusion at 4 weeks post transplantation (wpt). Blood vessel presence and maturation in the contusion at 1 wpt was similar in rats that received pMSC or untreated MSC. Nervous tissue sparing and functional recovery were similar across groups. Our results indicate that macrophage-derived inflammation priming does not increase the overall therapeutic potential of an MSC transplant in the adult rat contused spinal cord.
Collapse
|
65
|
Lymphotoxin β Receptor: a Crucial Role in Innate and Adaptive Immune Responses against Toxoplasma gondii. Infect Immun 2021; 89:IAI.00026-21. [PMID: 33753412 PMCID: PMC8316152 DOI: 10.1128/iai.00026-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 11/21/2022] Open
Abstract
The lymphotoxin β receptor (LTβR) plays an essential role in the initiation of immune responses to intracellular pathogens. In mice, the LTβR is crucial for surviving acute toxoplasmosis; however, until now, a functional analysis was largely incomplete. Here, we demonstrate that the LTβR is a key regulator required for the intricate balance of adaptive immune responses. Toxoplasma gondii-infected LTβR-deficient (LTβR−/−) mice show globally altered interferon-γ (IFN-γ) regulation, reduced IFN-γ-controlled host effector molecule expression, impaired T cell functionality, and an absent anti-parasite-specific IgG response, resulting in a severe loss of immune control of the parasites. Reconstitution of LTβR−/− mice with toxoplasma immune serum significantly prolongs survival following T. gondii infection. Notably, analysis of RNA-seq data clearly indicates a specific effect of T. gondii infection on the B cell response and isotype switching. This study uncovers the decisive role of the LTβR in cytokine regulation and adaptive immune responses to control T. gondii.
Collapse
|
66
|
Wilburn WJ, Jamal S, Ismail F, Brooks D, Whalen M. Evaluation of triclosan exposures on secretion of pro-inflammatory cytokines from human immune cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 83:103599. [PMID: 33516901 PMCID: PMC7956230 DOI: 10.1016/j.etap.2021.103599] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 05/06/2023]
Abstract
Triclosan (TCS) is widely used in personal hygiene products, such as mouthwash and toothpaste, and is found in human tissues. Interleukin (IL)-1 beta (IL-1β), IL-6, tumor necrosis factor alpha (TNFα), and interferon gamma (IFNγ) are pro-inflammatory cytokines and inappropriately elevated levels of each have been associated with pathologies including rheumatoid arthritis and certain cancers. Here we examine effects of TCS on the secretion of the pro-inflammatory cytokines from human immune cell preparations. TCS at concentrations between 0.05-5 μM consistently increased the secretion of IL-1β, IL-6, and TNFα within 24 h of exposure and the increases often maintained out to 6 days of exposure. TCS also induced increases in IFNγ secretion, however the increases were most consistent after 48 h of exposure rather than within 24 h. Additionally, a role for both p44/42 and p38 MAPK in TCS-stimulated increases in IL-1β was seen in cells from some donors.
Collapse
Affiliation(s)
- Wendy J Wilburn
- Departments of Biological Sciences, Tennessee State UnIversity, Nashville, TN, 37209, United States
| | - Sara Jamal
- Department of Chemistry, Tennessee State University, Nashville, TN, 37209, United States
| | - Farah Ismail
- Department of Chemistry, Tennessee State University, Nashville, TN, 37209, United States
| | - Dylan Brooks
- Department of Chemistry, Tennessee State University, Nashville, TN, 37209, United States
| | - Margaret Whalen
- Department of Chemistry, Tennessee State University, Nashville, TN, 37209, United States.
| |
Collapse
|
67
|
Beno SM, Riegler AN, Gilley RP, Brissac T, Wang Y, Kruckow KL, Jadapalli JK, Wright GM, Shenoy AT, Stoner SN, Restrepo MI, Deshane JS, Halade GV, González-Juarbe N, Orihuela CJ. Inhibition of Necroptosis to Prevent Long-term Cardiac Damage During Pneumococcal Pneumonia and Invasive Disease. J Infect Dis 2021; 222:1882-1893. [PMID: 32492702 DOI: 10.1093/infdis/jiaa295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Streptococcus pneumoniae infection can result in bacteremia with devastating consequences including heart damage. Necroptosis is a proinflammatory form of cell death instigated by pore-forming toxins such as S. pneumoniae pneumolysin. Necroptosis-inhibiting drugs may lessen organ damage during invasive pneumococcal disease (IPD). METHODS In vitro experiments were carried out with human and mouse cardiomyocytes. Long-term cardiac damage was assessed using high-resolution echocardiography in ampicillin-rescued mice 3 months after challenge with S. pneumoniae. Ponatinib, a necroptosis-inhibiting and Food and Drug Administration-approved drug for lymphocytic leukemia treatment, was administered intraperitoneally alongside ampicillin to test its therapeutic efficacy. Histology of heart sections included hematoxylin-eosin staining for overt damage, immunofluorescence for necroptosis, and Sirius red/fast green staining for collagen deposition. RESULTS Cardiomyocyte death and heart damage was due to pneumolysin-mediated necroptosis. IPD leads to long-term cardiac damage, as evidenced by de novo collagen deposition in mouse hearts and a decrease in fractional shortening. Adjunct necroptosis inhibition reduced the number of S. pneumoniae foci observed in hearts of acutely infected mice and serum levels of troponin I. Ponatinib reduced collagen deposition and protected heart function in convalescence. CONCLUSIONS Acute and long-term cardiac damage incurred during IPD is due in part to cardiomyocyte necroptosis. Necroptosis inhibitors may be a viable adjunct therapy.
Collapse
Affiliation(s)
- Sarah M Beno
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ashleigh N Riegler
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ryan P Gilley
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Terry Brissac
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yong Wang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Katherine L Kruckow
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeevan K Jadapalli
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Griffin M Wright
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anukul T Shenoy
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara N Stoner
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Jessy S Deshane
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ganesh V Halade
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Carlos J Orihuela
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
68
|
Liu X, Weng X, Xiao W, Xu X, Chen Y, Chen P. Pharmacological and Genetic Inhibition of PD-1 Demonstrate an Important Role of PD-1 in Ischemia-Induced Skeletal Muscle Inflammation, Oxidative Stress, and Angiogenesis. Front Immunol 2021; 12:586429. [PMID: 33815358 PMCID: PMC8017157 DOI: 10.3389/fimmu.2021.586429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/02/2021] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis is an important process under both physiological and pathophysiological conditions. Here we investigated the role and the underlying mechanism of PD-1 in hindlimb ischemia-induced inflammation and angiogenesis in mice. We found that inhibition of PD-1 by genetic PD-1 knockout or pharmacological PD-1 blocking antibodies dramatically attenuated hindlimb blood perfusion, angiogenesis, and exercise capacity in mice after femoral artery ligation. Mechanistically, we found that PD-1 knockout significantly exacerbated ischemia-induced muscle oxidative stress, leukocyte infiltration and IFN-γ production before abnormal angiogenesis in these mice. In addition, we found that the percentages of IFN-γ positive macrophages and CD8 T cells were significantly increased in P-1 knockout mice after hindlimb ischemia. Macrophages were the major leukocyte subset infiltrated in skeletal muscle, which were responsible for the enhanced muscle leukocyte-derived IFN-γ production in PD-1 knockout mice after hindlimb ischemia. Moreover, we demonstrated that IFN-γ significantly attenuated vascular endothelial cell proliferation, tube formation and migration in vitro. IFN-γ also significantly enhanced vascular endothelial cell apoptosis. In addition, the total number of TNF-α positive leukocytes/muscle weight were significantly increased in PD-1-/- mice after hindlimb ischemia. These data indicate that PD-1 exerts an important role in ischemia-induced muscle inflammation and angiogenesis.
Collapse
Affiliation(s)
- Xiaoguang Liu
- College of Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Xinyu Weng
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, United States.,Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yingjie Chen
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, United States.,Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
69
|
Accogli T, Bruchard M, Végran F. Modulation of CD4 T Cell Response According to Tumor Cytokine Microenvironment. Cancers (Basel) 2021; 13:cancers13030373. [PMID: 33498483 PMCID: PMC7864169 DOI: 10.3390/cancers13030373] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/08/2021] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
The advancement of knowledge on tumor biology over the past decades has demonstrated a close link between tumor cells and cells of the immune system. In this context, cytokines have a major role because they act as intermediaries in the communication into the tumor bed. Cytokines play an important role in the homeostasis of innate and adaptive immunity. In particular, they participate in the differentiation of CD4 T lymphocytes. These cells play essential functions in the anti-tumor immune response but can also be corrupted by tumors. The differentiation of naïve CD4 T cells depends on the cytokine environment in which they are activated. Additionally, at the tumor site, their activity can also be modulated according to the cytokines of the tumor microenvironment. Thus, polarized CD4 T lymphocytes can see their phenotype evolve, demonstrating functional plasticity. Knowledge of the impact of these cytokines on the functions of CD4 T cells is currently a source of innovation, for therapeutic purposes. In this review, we discuss the impact of the major cytokines present in tumors on CD4 T cells. In addition, we summarize the main therapeutic strategies that can modulate the CD4 response through their impact on cytokine production.
Collapse
Affiliation(s)
- Théo Accogli
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, 21000 Dijon, France; (T.A.); (M.B.)
- Team “CAdIR”, CRI INSERM UMR1231 “Lipids, Nutrition and Cancer”, Dijon 21000, France
- LipSTIC LabEx, 21000 Dijon, France
| | - Mélanie Bruchard
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, 21000 Dijon, France; (T.A.); (M.B.)
- Team “CAdIR”, CRI INSERM UMR1231 “Lipids, Nutrition and Cancer”, Dijon 21000, France
- LipSTIC LabEx, 21000 Dijon, France
- Centre Georges François Leclerc, 21000 Dijon, France
| | - Frédérique Végran
- Faculté des Sciences de Santé, Université Bourgogne Franche-Comté, 21000 Dijon, France; (T.A.); (M.B.)
- Team “CAdIR”, CRI INSERM UMR1231 “Lipids, Nutrition and Cancer”, Dijon 21000, France
- LipSTIC LabEx, 21000 Dijon, France
- Centre Georges François Leclerc, 21000 Dijon, France
- Correspondence:
| |
Collapse
|
70
|
Nazareth L, Walkden H, Chacko A, Delbaz A, Shelper T, Armitage CW, Reshamwala R, Trim LK, St John JA, Beagley KW, Ekberg JAK. Chlamydia muridarum Can Invade the Central Nervous System via the Olfactory and Trigeminal Nerves and Infect Peripheral Nerve Glial Cells. Front Cell Infect Microbiol 2021; 10:607779. [PMID: 33489937 PMCID: PMC7819965 DOI: 10.3389/fcimb.2020.607779] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Chlamydia pneumoniae can infect the brain and has been linked to late-onset dementia. Chlamydia muridarum, which infects mice, is often used to model human chlamydial infections. While it has been suggested to be also important for modelling brain infection, nervous system infection by C. muridarum has not been reported in the literature. C. pneumoniae has been shown to infect the olfactory bulb in mice after intranasal inoculation, and has therefore been suggested to invade the brain via the olfactory nerve; however, nerve infection has not been shown to date. Another path by which certain bacteria can reach the brain is via the trigeminal nerve, but it remains unknown whether Chlamydia species can infect this nerve. Other bacteria that can invade the brain via the olfactory and/or trigeminal nerve can do so rapidly, however, whether Chlamydia spp. can reach the brain earlier than one-week post inoculation remains unknown. In the current study, we showed that C. muridarum can within 48 h invade the brain via the olfactory nerve, in addition to infecting the trigeminal nerve. We also cultured the glial cells of the olfactory and trigeminal nerves and showed that C. muridarum readily infected the cells, constituting a possible cellular mechanism explaining how the bacteria can invade the nerves without being eliminated by glial immune functions. Further, we demonstrated that olfactory and trigeminal glia differed in their responses to C. muridarum, with olfactory glia showing less infection and stronger immune response than trigeminal glia.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Heidi Walkden
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Anu Chacko
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Ali Delbaz
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Todd Shelper
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Charles W Armitage
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ronak Reshamwala
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Logan K Trim
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - James A St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Kenneth W Beagley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jenny A K Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
71
|
Ma L, Gonzalez-Junca A, Zheng Y, Ouyang H, Illa-Bochaca I, Horst KC, Krings G, Wang Y, Fernandez-Garcia I, Chou W, Barcellos-Hoff MH. Inflammation Mediates the Development of Aggressive Breast Cancer Following Radiotherapy. Clin Cancer Res 2021; 27:1778-1791. [PMID: 33402361 DOI: 10.1158/1078-0432.ccr-20-3215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/23/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Women treated with radiotherapy before 30 years of age have increased risk of developing breast cancer at an early age. Here, we sought to investigate mechanisms by which radiation promotes aggressive cancer. EXPERIMENTAL DESIGN The tumor microenvironment (TME) of breast cancers arising in women treated with radiotherapy for Hodgkin lymphoma was compared with that of sporadic breast cancers. To investigate radiation effects on carcinogenesis, we analyzed tumors arising from Trp53-null mammary transplants after irradiation of the target epithelium or host using immunocompetent and incompetent mice, some of which were treated with aspirin. RESULTS Compared with age-matched specimens of sporadic breast cancer, radiation-preceded breast cancers (RP-BC) were characterized by TME rich in TGFβ, cyclooxygenase 2, and myeloid cells, indicative of greater immunosuppression, even when matched for triple-negative status. The mechanism by which radiation impacts TME construction was investigated in carcinomas arising in mice bearing Trp53-null mammary transplants. Immunosuppressive TMEs (iTME) were recapitulated in mice irradiated before transplantation, which implicated systemic immune effects. In nu/nu mice lacking adaptive immunity irradiated before Trp53-null mammary transplantation, cancers also established an iTME, which pointed to a critical role for myeloid cells. Consistent with this, irradiated mammary glands contained more macrophages and human cells cocultured with polarized macrophages underwent dysplastic morphogenesis mediated by IFNγ. Treating mice with low-dose aspirin for 6 months postirradiation prevented establishment of an iTME and resulted in less aggressive tumors. CONCLUSIONS These data show that radiation acts via nonmutational mechanisms to promote markedly immunosuppressive features of aggressive, RP-BCs.
Collapse
Affiliation(s)
- Lin Ma
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Alba Gonzalez-Junca
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Yufei Zheng
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Haoxu Ouyang
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
| | - Irineu Illa-Bochaca
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
| | - Kathleen C Horst
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Yinghao Wang
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | | | - William Chou
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
72
|
The Antimicrobial Peptide, Bactenecin 5, Supports Cell-Mediated but Not Humoral Immunity in the Context of a Mycobacterial Antigen Vaccine Model. Antibiotics (Basel) 2020; 9:antibiotics9120926. [PMID: 33352656 PMCID: PMC7766334 DOI: 10.3390/antibiotics9120926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 01/28/2023] Open
Abstract
Bactenecin (Bac) 5 is a bovine antimicrobial peptide (AMP) capable of killing some species of bacteria through the inhibition of protein synthesis. Bac5 and other AMPs have also been shown to have chemotactic properties and can induce inflammatory cytokine expression by innate immune cells. Recently, AMPs have begun to be investigated for their potential use as novel vaccine adjuvants. In the current work, we characterise the functionality of Bac5 in vitro using murine macrophage-like cells, ex vivo using human tonsil tissue and in vivo using a murine model of vaccination. We report the effects of the peptide in isolation and in the context of co-presentation with mycobacterial antigen and whole, inert Bacillus subtilis spore antigens. We find that Bac5 can trigger the release of nitric oxide from murine macrophages and upregulate surface marker expression including CD86, MHC-I and MHC-II, in the absence of additional agonists. When coupled with mycobacterial Ag85 and B. subtilis spores, Bac5 also enhanced IFNγ secretion. We provide evidence that B. subtilis spores, but not the Bac5 peptide, act as strong adjuvants in promoting antigen-specific immunoglobulin production in Ag85B-vaccinated mice. Our findings suggest that Bac5 is an important regulator of the early cell-mediated host immune response.
Collapse
|
73
|
Kirshtein A, Akbarinejad S, Hao W, Le T, Su S, Aronow RA, Shahriyari L. Data Driven Mathematical Model of Colon Cancer Progression. J Clin Med 2020; 9:E3947. [PMID: 33291412 PMCID: PMC7762015 DOI: 10.3390/jcm9123947] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Every colon cancer has its own unique characteristics, and therefore may respond differently to identical treatments. Here, we develop a data driven mathematical model for the interaction network of key components of immune microenvironment in colon cancer. We estimate the relative abundance of each immune cell from gene expression profiles of tumors, and group patients based on their immune patterns. Then we compare the tumor sensitivity and progression in each of these groups of patients, and observe differences in the patterns of tumor growth between the groups. For instance, in tumors with a smaller density of naive macrophages than activated macrophages, a higher activation rate of macrophages leads to an increase in cancer cell density, demonstrating a negative effect of macrophages. Other tumors however, exhibit an opposite trend, showing a positive effect of macrophages in controlling tumor size. Although the results indicate that for all patients the size of the tumor is sensitive to the parameters related to macrophages, such as their activation and death rate, this research demonstrates that no single biomarker could predict the dynamics of tumors.
Collapse
Affiliation(s)
- Arkadz Kirshtein
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003-9305, USA; (A.K.); (S.A.); (T.L.); (S.S.); (R.A.A.)
| | - Shaya Akbarinejad
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003-9305, USA; (A.K.); (S.A.); (T.L.); (S.S.); (R.A.A.)
| | - Wenrui Hao
- Department of Mathematics, Pennsylvania State University, University Park, State College, PA 16802, USA;
| | - Trang Le
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003-9305, USA; (A.K.); (S.A.); (T.L.); (S.S.); (R.A.A.)
| | - Sumeyye Su
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003-9305, USA; (A.K.); (S.A.); (T.L.); (S.S.); (R.A.A.)
| | - Rachel A. Aronow
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003-9305, USA; (A.K.); (S.A.); (T.L.); (S.S.); (R.A.A.)
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003-9305, USA; (A.K.); (S.A.); (T.L.); (S.S.); (R.A.A.)
| |
Collapse
|
74
|
Jundi D, Krayem I, Bazzi S, Karam M. In vitro effects of azide-containing human CRP isoforms and oxLDL on U937-derived macrophage production of atherosclerosis-related cytokines. Exp Ther Med 2020; 20:57. [PMID: 32952647 DOI: 10.3892/etm.2020.9185] [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: 07/14/2019] [Accepted: 11/18/2019] [Indexed: 11/05/2022] Open
Abstract
Atherosclerosis is an inflammatory chronic disease of the arterial wall. Monomeric (m) and pentameric (p) C-reactive protein (CRP) and oxidized low density lipoproteins (oxLDL) seem to affect the pattern of cytokine production by macrophages, thus playing an important role in atherogenesis. Azide, the commercial preservative of CRP, may influence its action in vitro. The present study aimed to determine the effects of both isoforms of azide-containing CRP (mCRP and pCRP) with and without oxLDL on cytokine production by U937-derived macrophages. U937 monocytes were cultured and differentiated into macrophages and treated with mCRP, pCRP, oxLDL and azide individually and in combination. ELISA were performed to measure the levels of interferon-γ (IFN-γ), interleukin (IL)-4, IL-6, IL-10 and tumor necrosis factor (TNF)-α in culture supernatants collected from U937-derived macrophages following their respective treatments. Most single and combined treatments, especially in triple combination, were able to downregulate the levels of IFN-γ and IL-6 compared with control untreated cells, whilst the combination of mCRP and pCRP increased IL-4 levels. Regarding IL-10, except for an increase induced by mCRP, no significant effect was caused by any treatment compared with the control. On the other hand, the levels of TNF-α were not significantly affected by any treatment except for a decreasing trend that was observed with mCRP/oxLDL treatment compared with control. By contrast, double azide caused a significant decrease in the levels of IFN-γ and IL-6. The results of the present study indicated that mCRP, pCRP, oxLD and possibly azide, individually or in different combinations, had the tendency to upregulate the expression of IL-4 and to downregulate that of the pro-atherogenic cytokines, IFN-γ and IL-6, suggesting that the intima microenvironment serves a crucial role in atherogenesis.
Collapse
Affiliation(s)
- Dania Jundi
- Department of Biology, University of Balamand, Kourah, P. O. Box 100 Tripoli, North Governorate, Lebanon
| | - Imtissal Krayem
- Department of Biology, University of Balamand, Kourah, P. O. Box 100 Tripoli, North Governorate, Lebanon
| | - Samer Bazzi
- Department of Biology, University of Balamand, Kourah, P. O. Box 100 Tripoli, North Governorate, Lebanon
| | - Marc Karam
- Department of Biology, University of Balamand, Kourah, P. O. Box 100 Tripoli, North Governorate, Lebanon
| |
Collapse
|
75
|
Li R, Ying B, Liu Y, Spencer JF, Miao J, Tollefson AE, Brien JD, Wang Y, Wold WSM, Wang Z, Toth K. Generation and characterization of an Il2rg knockout Syrian hamster model for XSCID and HAdV-C6 infection in immunocompromised patients. Dis Model Mech 2020; 13:dmm044602. [PMID: 32651192 PMCID: PMC7473636 DOI: 10.1242/dmm.044602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
Model animals are indispensable for the study of human diseases, and in general, of complex biological processes. The Syrian hamster is an important model animal for infectious diseases, behavioral science and metabolic science, for which more experimental tools are becoming available. Here, we describe the generation and characterization of an interleukin-2 receptor subunit gamma (Il2rg) knockout (KO) Syrian hamster strain. In humans, mutations in IL2RG can result in a total failure of T and natural killer (NK) lymphocyte development and nonfunctional B lymphocytes (X-linked severe combined immunodeficiency; XSCID). Therefore, we sought to develop a non-murine model to study XSCID and the infectious diseases associated with IL2RG deficiency. We demonstrated that the Il2rg KO hamsters have a lymphoid compartment that is greatly reduced in size and diversity, and is impaired in function. As a result of the defective adaptive immune response, Il2rg KO hamsters developed a more severe human adenovirus infection and cleared virus less efficiently than immune competent wild-type hamsters. Because of this enhanced virus replication, Il2rg KO hamsters developed more severe adenovirus-induced liver pathology than wild-type hamsters. This novel hamster strain will provide researchers with a new tool to investigate human XSCID and its related infections.
Collapse
Affiliation(s)
- Rong Li
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Yanan Liu
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
- National Center for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Yaohe Wang
- National Center for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - William S M Wold
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| |
Collapse
|
76
|
Xu B, Liu R, Ding M, Zhang J, Sun H, Liu C, Lu F, Zhao S, Pan Q, Zhang X. Interaction of Mycoplasma synoviae with chicken synovial sheath cells contributes to macrophage recruitment and inflammation. Poult Sci 2020; 99:5366-5377. [PMID: 33142453 PMCID: PMC7647830 DOI: 10.1016/j.psj.2020.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/13/2020] [Accepted: 08/15/2020] [Indexed: 11/21/2022] Open
Abstract
Mycoplasma synoviae (MS) is an important avian pathogen causing considerable economic hardship in the poultry industry. A major inflammation caused by MS is synovitis that occurs in the synovial tendon sheath and joint synovium. However, the overall appearance of pathological changes in the tendon sheath and surrounding tissues caused by MS infection at the level of pathological tissue sections was poor. Studies on the role of MS and synovial sheath cells (SSCs) interaction in the development of synovitis have not been carried out. Through histopathological observation, our study found that a major MS-induced pathological change of the tendon sheath synovium was extensive scattered and focal inflammatory cell infiltration of the tendon sheath synovial layer. In vitro research experiments revealed that the CFU numbers of MS adherent and invading SSC, the levels of expression of various pattern recognition receptors, inflammatory cytokines, and chemokines coding genes, such as IL-1β, IL-6, IL-8, CCL-20, RANTES, MIP-1β, TLR7, and TLR15 in SSCs, and chemotaxis of macrophages were significantly increased when the multiplicity of infection (MOI) of MS to SSC were increased tenfold. The expression level of IL-12p40 in SSC was significantly higher when the MOIs of MS to SSC were increased by a factor of 100. The interaction between MS and SSC can activate macrophages, which was manifested by a significant increase in the expression of IL-1β, IL-6, IL-8, CCL-20, RANTES, MIP-1β, and CXCL-13. This study systematically demonstrated that the interaction of MS with chicken SSC contributes to the inflammatory response caused by the robust expression of related cytokines and macrophage chemotaxis. These findings are helpful in elucidating the molecular mechanism of MS-induced synovitis in chickens.
Collapse
Affiliation(s)
- Bin Xu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Rui Liu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Meijuan Ding
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jingfeng Zhang
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Huawei Sun
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Chuanmin Liu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Fengying Lu
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Sha Zhao
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qunxing Pan
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Xiaofei Zhang
- Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; National Center for Engineering Research of Veterinary Bio-Products, Jiangsu Academy of Agricultural Sciences, Nanjing, China.
| |
Collapse
|
77
|
Norbury AJ, Calvert JK, Al-Shujairi WH, Cabezas-Falcon S, Tang V, Ong LC, Alonso SL, Smith JR, Carr JM. Dengue virus infects the mouse eye following systemic or intracranial infection and induces inflammatory responses. J Gen Virol 2020; 101:79-85. [PMID: 31774391 DOI: 10.1099/jgv.0.001354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dengue virus (DENV) infection is associated with clinical ocular presentations and here DENV infection of the eye was assessed in mice. In an AG129 mouse model of antibody-dependent enhancement of DENV infection, DENV RNA was detected in the eye and vascular changes were present in the retinae. Intraocular CD8 and IFN-γ mRNA were increased in mice born to DENV-naïve, but not DENV-immune mothers, while TNF-α mRNA was induced and significantly higher in mice born to DENV-immune than DENV-naïve mothers. DENV RNA was detected in the eye following intracranial DENV infection and CD8 mRNA but not IFN-γ nor TNF-α were induced. In all models, viperin was increased following DENV infection. Thus, DENV in the circulation or the brain can infect the eye and stimulate innate immune responses, with induction of viperin as one response that consistently occurs in multiple DENV eye-infection models in both an IFN-dependent and independent manner.
Collapse
Affiliation(s)
- Aidan J Norbury
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Julie K Calvert
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Wisam H Al-Shujairi
- Department of Laboratory and Clinical Sciences, College of Pharmacy, University of Babylon, Hilla 51002, Iraq.,Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Sheila Cabezas-Falcon
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Victoria Tang
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Li Ching Ong
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and Immunology programme, Life Sciences Institute, National University of Singapore, 21 Lower Kent Ridge Rd, Singapore
| | - Sylvie L Alonso
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and Immunology programme, Life Sciences Institute, National University of Singapore, 21 Lower Kent Ridge Rd, Singapore
| | - Justine R Smith
- Eye and Vision Health, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Jillian M Carr
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| |
Collapse
|
78
|
Ibrahim S, Harris-Kawano A, Haider I, Mirmira RG, Sims EK, Anderson RM. A novel Cre-enabled tetracycline-inducible transgenic system for tissue-specific cytokine expression in the zebrafish: CETI-PIC3. Dis Model Mech 2020; 13:dmm042556. [PMID: 32457041 PMCID: PMC7328138 DOI: 10.1242/dmm.042556] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/17/2020] [Indexed: 01/21/2023] Open
Abstract
Maladaptive signaling by pro-inflammatory cytokines (PICs), such as TNFα, IL1β and IFNɣ, can activate downstream signaling cascades that are implicated in the development and progression of multiple inflammatory diseases. Despite playing critical roles in pathogenesis, the availability of in vivo models in which to model tissue-specific induction of PICs is limited. To bridge this gap, we have developed a novel multi-gene expression system dubbed Cre-enabled and tetracycline-inducible transgenic system for conditional, tissue-specific expression of pro-inflammatory cytokines (CETI-PIC3). This binary transgenic system permits the stoichiometric co-expression of proteins Tumor necrosis factor a (Tnfa), Interleukin-1 beta (Il1b) and Interferon gamma (Ifng1), and H2B-GFP fluorescent reporter in a dose-dependent manner. Furthermore, cytokine misexpression is enabled only in tissue domains that can be defined by Cre recombinase expression. We have validated this system in zebrafish using an insulin:cre line. In doubly transgenic fish, quantitative real-time polymerase chain reaction demonstrated increased expression levels of tnfa, il1b and ifng1 mRNA. Moreover, specific expression in pancreatic β cells was demonstrated by both Tnfa immunofluorescence and GFP fluorescence. Cytokine-overexpressing islets elicited specific responses: β cells exhibited increased expression of genes associated with reactive oxidative species-mediated stress and endoplasmic reticulum stress, surveilling and infiltrating macrophages were increased, and β cell death was promoted. This powerful and versatile model system can be used for modeling, analysis and therapy development of diseases with an underlying inflammatory etiology.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sara Ibrahim
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Arianna Harris-Kawano
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Isra Haider
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G Mirmira
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Emily K Sims
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ryan M Anderson
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
79
|
Mezouar S, Mege JL. Changing the paradigm of IFN-γ at the interface between innate and adaptive immunity: Macrophage-derived IFN-γ. J Leukoc Biol 2020; 108:419-426. [PMID: 32531848 DOI: 10.1002/jlb.4mir0420-619rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
IFN-γ plays a critical role in the immune response to bacterial infections. It is established that IFN-γ is mainly produced by NK/ILC1 cells and T cells, and most of papers have rejected the biologic reality of alternative sources for more than 20 years. Here, we are proposing to revisit this dogma and discuss the role of macrophage-derived IFN-γ in bacterial infections. Our hypothesis is based on a panel of publications and is recently revived by our results on placenta, a chimeric organ in which the immune response is tailored to protect the fetus from mother's immune response. The culture of purified placental macrophages is associated with a production of IFN-γ that may contribute to fetal protection from bacterial infections before eliciting a Th1-like immune response potentially pathogenic for pregnancy. Hence, macrophage IFN-γ may be a novel actor of early crosstalk between innate and adaptive immunity in the context of host defense against bacterial infections.
Collapse
Affiliation(s)
- Soraya Mezouar
- Aix-Marseille University, MEPHI, IRD, APHM, Marseille, France.,IHU-Mediterranean Infection, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille University, MEPHI, IRD, APHM, Marseille, France.,IHU-Mediterranean Infection, Marseille, France.,APHM, UF Immunology Department, Marseille, France
| |
Collapse
|
80
|
KSHV infection skews macrophage polarisation towards M2-like/TAM and activates Ire1 α-XBP1 axis up-regulating pro-tumorigenic cytokine release and PD-L1 expression. Br J Cancer 2020; 123:298-306. [PMID: 32418990 PMCID: PMC7374093 DOI: 10.1038/s41416-020-0872-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/27/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022] Open
Abstract
Background Kaposi’s Sarcoma Herpesvirus (KSHV) is a gammaherpesvirus strongly linked to human cancer. The virus is also able to induce immune suppression, effect that contributes to onset/progression of the viral-associated malignancies. As KSHV may infect macrophages and these cells abundantly infiltrate Kaposi’s sarcoma lesions, in this study we investigated whether KSHV-infection could affect macrophage polarisation to promote tumorigenesis. Methods FACS analysis was used to detect macrophage markers and PD-L1 expression. KSHV infection and the molecular pathways activated were investigated by western blot analysis and by qRT-PCR while cytokine release was assessed by Multi-analyte Kit. Results We found that KSHV infection reduced macrophage survival and skewed their polarisation towards M2 like/TAM cells, based on the expression of CD163, on the activation of STAT3 and STAT6 pathways and the release of pro-tumorigenic cytokines such as IL-10, VEGF, IL-6 and IL-8. We also found that KSHV triggered Ire1 α-XBP1 axis activation in infected macrophages to increase the release of pro-tumorigenic cytokines and to up-regulate PD-L1 surface expression. Conclusions The findings that KSHV infection of macrophages skews their polarisation towards M2/TAM and that activate Ire1 α-XBP1 to increase the release of pro-tumorigenic cytokines and the expression of PD-L1, suggest that manipulation of UPR could be exploited to prevent or improve the treatment of KSHV-associated malignancies.
Collapse
|
81
|
Guerrero-Arguero I, Høj TR, Tass ES, Berges BK, Robison RA. A comparison of Chikungunya virus infection, progression, and cytokine profiles in human PMA-differentiated U937 and murine RAW264.7 monocyte derived macrophages. PLoS One 2020; 15:e0230328. [PMID: 32163514 PMCID: PMC7067478 DOI: 10.1371/journal.pone.0230328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/27/2020] [Indexed: 11/29/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes rash, fever and severe polyarthritis that can last for years in humans. Murine models display inflammation and macrophage infiltration only in the adjacent tissues at the site of inoculation, showing no signs of systemic polyarthritis. Monocyte-derived macrophages are one cell type suspected to contribute to a systemic CHIKV infection. The purpose of this study was to analyze differences in CHIKV infection in two different cell lines, human U937 and murine RAW264.7 monocyte derived macrophages. PMA-differentiated U937 and RAW264.7 macrophages were infected with CHIKV, and infectious virus production was measured by plaque assay and by reverse transcriptase quantitative PCR at various time points. Secreted cytokines in the supernatants were measured using cytometric bead arrays. Cytokine mRNA levels were also measured to supplement expression data. Here we show that CHIKV replicates more efficiently in human macrophages compared to murine macrophages. In addition, infected human macrophages produced around 10-fold higher levels of infectious virus when compared to murine macrophages. Cytokine induction by CHIKV infection differed between human and murine macrophages; IL-1, IL-6, IFN-γ, and TNF were significantly upregulated in human macrophages. This evidence suggests that CHIKV replicates more efficiently and induces a much greater pro-inflammatory cytokine profile in human macrophages, when compared to murine macrophages. This may shed light on the critical role that macrophages play in the CHIKV inflammatory response.
Collapse
Affiliation(s)
- Israel Guerrero-Arguero
- Department of Microbiology and Molecular Biology, College of Life Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Taalin R. Høj
- Department of Microbiology and Molecular Biology, College of Life Sciences, Brigham Young University, Provo, Utah, United States of America
| | - E. Shannon Tass
- Department of Statistics, College of Physical and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Bradford K. Berges
- Department of Microbiology and Molecular Biology, College of Life Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Richard A. Robison
- Department of Microbiology and Molecular Biology, College of Life Sciences, Brigham Young University, Provo, Utah, United States of America
| |
Collapse
|
82
|
Sanderford V, Barna BP, Barrington RA, Malur A, Mohan A, Leffler N, Soliman E, Thomassen MJ. PPARγ Deficiency in Carbon Nanotube-elicited Granulomatous Inflammation Promotes a Th17 Response to a Microbial Antigen. JOURNAL OF NANOMEDICINE & NANOTECHNOLOGY 2020; 11:541. [PMID: 32405439 PMCID: PMC7219023 DOI: 10.35248/2157-7439.20.11.541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The pathological consequences of interaction between environmental carbon pollutants and microbial antigens have not been fully explored. We developed a murine model of multi-wall carbon nanotube (MWCNT)-elicited granulomatous disease which bears a striking resemblance to sarcoidosis, a human granulomatous disease. Because of reports describing lymphocyte reactivity to mycobacterial antigens in sarcoidosis patients, we hypothesized that addition of mycobacterial antigen (ESAT-6) to MWCNT might elicit activation in T cells. METHODS Macrophage-specific peroxisome-proliferator-activated receptor gamma (PPARγ) knock out (KO) mice were studied along with wild-type mice because our previous report indicated PPARγ deficiency in sarcoidosis alveolar macrophages. MWCNT+ESAT-6 were instilled into mice. Controls received vehicle (surfactant-PBS) or ESAT-6 and were evaluated 60 days post-instillation. As noted in our recent publication, lung tissues from PPARγ KO mice instilled with MWCNT+ESAT-6 yielded more intensive pathophysiology, with elevated fibrosis. RESULTS Inspection of mediastinal lymph nodes (MLN) revealed no granulomas but deposition of MWCNT. MLN cell counts were higher in PPARγ KO than in wild-type instilled with MWCNT+ESAT-6. Moreover, the CD4:CD8 T cell ratio, a major clinical metric for human disease, was increased in PPARγ KO mice. Bronchoalveolar lavage (BAL) cells from PPARγ KO mice instilled with MWCNT+ESAT-6 displayed increased Th17 cell markers (RORγt, IL-17A, CCR6) which associate with elevated fibrosis. CONCLUSION These findings suggest that PPARγ deficiency in macrophages may promote ESAT-6-associated T cell activation in the lung, and that the MWCNT+ESAT-6 model may offer new insights into pathways of lymphocyte-mediated sarcoidosis histopathology.
Collapse
Affiliation(s)
- Victoria Sanderford
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Barbara P Barna
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Robert A Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Anagha Malur
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Arjun Mohan
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Nancy Leffler
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Eman Soliman
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mary Jane Thomassen
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| |
Collapse
|
83
|
Alterations in the innate immune system due to exhausting exercise in intensively trained rats. Sci Rep 2020; 10:967. [PMID: 31969634 PMCID: PMC6976645 DOI: 10.1038/s41598-020-57783-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 11/11/2019] [Indexed: 01/10/2023] Open
Abstract
It is known that intensive physical activity alters the immune system's functionality. However, the influence of the intensity and duration of exercise needs to be studied in more depth. We aimed to establish the changes in the innate immune response induced by two programmes of intensive training in rats compared to sedentary rats. A short training programme included 2 weeks of intensive training, ending with an exhaustion test (short training with exhaustion, S-TE). A second training programme comprised 5-week training including two exhaustion tests and three trainings per week. In this case, immune status was assessed before (T), immediately after (TE) and 24 h after (TE24) an additional final exhaustion test. Biomarkers such as phagocytic activity, macrophage cytokine and reactive oxygen species (ROS) production, and natural killer (NK) cell activity were quantified. S-TE was not enough to induce changes in the assessed innate immunity biomarkers. However, the second training was accompanied by a decrease in the phagocytic activity, changes in the pattern of cytokine secretion and ROS production by macrophages and reduced NK cell proportion but increased NK cytotoxic activity. In conclusion, a 5-week intense training programme, but not a shorter training, induced alterations in the innate immune system functionality.
Collapse
|
84
|
Schneider A, Wood HN, Geden S, Greene CJ, Yates RM, Masternak MM, Rohde KH. Growth hormone-mediated reprogramming of macrophage transcriptome and effector functions. Sci Rep 2019; 9:19348. [PMID: 31852980 PMCID: PMC6920138 DOI: 10.1038/s41598-019-56017-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/29/2019] [Indexed: 01/07/2023] Open
Abstract
Macrophages are an important component of the innate immune response. Priming and activation of macrophages is stimulated by cytokines (i.e IFNγ). However, growth hormone (GH) can also stimulate macrophage activation. Based on these observations, the goal of this work was to 1) to compare the transcriptome profile of macrophages activated in vitro with GH and IFNγ, and 2) to assess the impact of GH on key macrophage functional properties like reactive oxygen species (ROS) production and phagosomal proteolysis. To assess the global transcriptional and functional impact of GH on macrophage programming, bone marrow derived macrophages were treated with GH or IFNγ. Our data strongly support a potential link between GH, which wanes with age, and impaired macrophage function. The notable overlap of GH with IFNγ-induced pathways involved in innate immune sensing of pathogens and antimicrobial responses argue for an important role for GH in macrophage priming and maturation. By using functional assays that report on biochemical activities within the lumen of phagosomes, we have also shown that GH alters physiologically relevant processes such as ROS production and proteolysis. These changes could have far reaching impacts on antimicrobial capacity, signaling, and antigen presentation.
Collapse
Affiliation(s)
- Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Hillary N Wood
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Sandra Geden
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Catherine J Greene
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA.
- Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland.
| | - Kyle H Rohde
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA.
| |
Collapse
|
85
|
Liu S, Karaganis S, Mo RF, Li XX, Wen RX, Song XJ. IFNβ Treatment Inhibits Nerve Injury-induced Mechanical Allodynia and MAPK Signaling By Activating ISG15 in Mouse Spinal Cord. THE JOURNAL OF PAIN 2019; 21:836-847. [PMID: 31785403 DOI: 10.1016/j.jpain.2019.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/15/2019] [Accepted: 11/11/2019] [Indexed: 01/10/2023]
Abstract
Neuropathic pain is difficult to treat and remains a major clinical challenge worldwide. While the mechanisms which underlie the development of neuropathic pain are incompletely understood, interferon signaling by the immune system is known to play a role. Here, we demonstrate a role for interferon β (IFNβ) in attenuating mechanical allodynia induced by the spared nerve injury in mice. The results show that intrathecal administration of IFNβ (dosages up to 5,000 U) produces significant, transient, and dose-dependent attenuation of mechanical allodynia without observable effects on motor activity or feeding behavior, as is common with IFN administration. This analgesic effect is mediated by the ubiquitin-like protein interferon-stimulated gene 15 (ISG15), which is potently induced within the spinal cord following intrathecal delivery of IFNβ. Both free and conjugated ISG15 are elevated following IFNβ treatment, and this effect is increased in UBP43-/- mice lacking a key deconjugating enzyme. The IFNβ-mediated analgesia reduces MAPK signaling activation following nerve injury, and this effect requires induction of ISG15. These findings highlight a new role for IFNβ, ISG15, and MAPK signaling in immunomodulation of neuropathic pain and may lead to new therapeutic possibilities. PERSPECTIVE: Neuropathic pain is frequently intractable in a clinical setting, and new treatment options are needed. Characterizing the antinociceptive potential of IFNβ and the associated downstream signaling pathways in preclinical models may lead to the development of new therapeutic options for debilitating neuropathies.
Collapse
Affiliation(s)
- Su Liu
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China; Department of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Stephen Karaganis
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China; Department of Life, Earth and Environmental Sciences, West Texas A&M University, Amarillo, Texas
| | - Ru-Fan Mo
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiao-Xiao Li
- Department of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruo-Xin Wen
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xue-Jun Song
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
86
|
Gaydos J, McNally A, Burnham EL. The impact of alcohol use disorders on pulmonary immune cell inflammatory responses to Streptococcus pneumoniae. Alcohol 2019; 80:119-130. [PMID: 30195043 DOI: 10.1016/j.alcohol.2018.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Community-acquired pneumonia due to Streptococcus pneumoniae occurs commonly in alcohol use disorders (AUDs). Pneumonia in the AUD patient is associated with poorer outcomes, and specific therapies to mitigate disease severity in these patients do not exist. Numerous investigations have attributed increased severity of pneumonia in AUDs to aberrant function of the alveolar macrophage (AM), a lung immune cell critical in host defense initiation. No studies have examined the response of human AMs to S. pneumoniae in AUDs. We hypothesized that the inflammatory mediators released by AMs after S. pneumoniae stimulation would differ quantitatively in individuals with AUDs compared to non-AUD participants. We further postulated that AM inflammatory mediators would be diminished after exposure to the antioxidant, N-acetylcysteine (NAC). For comparison, responses of peripheral blood mononuclear cells (PBMCs) to pneumococcal protein were also examined. Otherwise healthy participants with AUDs and smoking-matched controls underwent bronchoalveolar lavage and peripheral blood sampling to obtain AMs and PBMCs, respectively. Freshly collected cells were cultured with increasing doses of heat-killed S. pneumoniae protein, with and without exposure to N-acetylcysteine. Cell culture supernatants were collected, and inflammatory mediators were measured, including interferon (IFN)-γ, interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. IFN-γ and IL-6 were significantly higher in unstimulated AM cell culture supernatants from subjects with AUDs. After stimulation with pneumococcal protein, a dose-response and time-dependent increase in pro-inflammatory cytokine production by both AMs and PBMCs was also observed; differences were not observed between AUD and non-AUD subjects. Addition of NAC to pneumococcal-stimulated AMs and PBMCs was generally associated with diminished cytokine production, with the exception of IL-1β that was elevated in AM culture supernatants from subjects with AUDs. Our observations suggest that AUDs contribute to basal alterations in AM pro-inflammatory cytokine elaboration, but did not support consistent differences in pneumococcal-stimulated AM or PBMC inflammatory mediator secretion that were referable to AUDs.
Collapse
|
87
|
Tuladhar S, Kochanowsky JA, Bhaskara A, Ghotmi Y, Chandrasekaran S, Koshy AA. The ROP16III-dependent early immune response determines the subacute CNS immune response and type III Toxoplasma gondii survival. PLoS Pathog 2019; 15:e1007856. [PMID: 31648279 PMCID: PMC6812932 DOI: 10.1371/journal.ppat.1007856] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022] Open
Abstract
Toxoplasma gondii is an intracellular parasite that persistently infects the CNS and that has genetically distinct strains which provoke different acute immune responses. How differences in the acute immune response affect the CNS immune response is unknown. To address this question, we used two persistent Toxoplasma strains (type II and type III) and examined the CNS immune response at 21 days post infection (dpi). Contrary to acute infection studies, type III-infected mice had higher numbers of total CNS T cells and macrophages/microglia but fewer alternatively activated macrophages (M2s) and regulatory T cells (Tregs) than type II-infected mice. By profiling splenocytes at 5, 10, and 21 dpi, we determined that at 5 dpi type III-infected mice had more M2s while type II-infected mice had more pro-inflammatory macrophages and that these responses flipped over time. To test how these early differences influence the CNS immune response, we engineered the type III strain to lack ROP16 (IIIΔrop16), the polymorphic effector protein that drives the early type III-associated M2 response. IIIΔrop16-infected mice showed a type II-like neuroinflammatory response with fewer infiltrating T cells and macrophages/microglia and more M2s and an unexpectedly low CNS parasite burden. At 5 dpi, IIIΔrop16-infected mice showed a mixed inflammatory response with more pro-inflammatory macrophages, M2s, T effector cells, and Tregs, and decreased rates of infection of peritoneal exudative cells (PECs). These data suggested that type III parasites need the early ROP16-associated M2 response to avoid clearance, possibly by the Immunity-Related GTPases (IRGs), which are IFN-γ- dependent proteins essential for murine defenses against Toxoplasma. To test this possibility, we infected IRG-deficient mice and found that IIIΔrop16 parasites now maintained parental levels of PECs infection. Collectively, these studies suggest that, for the type III strain, rop16III plays a key role in parasite persistence and influences the subacute CNS immune response. Toxoplasma is a ubiquitous intracellular parasite that establishes an asymptomatic brain infection in immunocompetent individuals. However, in the immunocompromised and the developing fetus, Toxoplasma can cause problems ranging from fever to chorioretinitis to severe toxoplasmic encephalitis. Emerging evidence suggests that the genotype of the infecting Toxoplasma strain may influence these outcomes, possibly through the secretion of Toxoplasma strain-specific polymorphic effector proteins that trigger different host cell signaling pathways. While such strain-specific modulation of host cell signaling has been shown to affect acute immune responses, it is unclear how these differences influence the subacute or chronic responses in the CNS, the major organ affected in symptomatic disease. This study shows that genetically distinct strains of Toxoplasma provoke strain-specific CNS immune responses and that, for one strain (type III), acute and subacute immune responses and parasite survival are heavily influenced by a polymorphic parasite gene (rop16III).
Collapse
Affiliation(s)
- Shraddha Tuladhar
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Joshua A. Kochanowsky
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Apoorva Bhaskara
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Yarah Ghotmi
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Undergraduate Biology Research Program (UBRP), University of Arizona, Tucson, Arizona, United States of America
| | | | - Anita A. Koshy
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
88
|
Keuper M. On the role of macrophages in the control of adipocyte energy metabolism. Endocr Connect 2019; 8:R105-R121. [PMID: 31085768 PMCID: PMC6590200 DOI: 10.1530/ec-19-0016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022]
Abstract
The crosstalk between macrophages (MΦ) and adipocytes within white adipose tissue (WAT) influences obesity-associated insulin resistance and other associated metabolic disorders, such as atherosclerosis, hypertension and type 2 diabetes. MΦ infiltration is increased in WAT during obesity, which is linked to decreased mitochondrial content and activity. The mechanistic interplay between MΦ and mitochondrial function of adipocytes is under intense investigation, as MΦ and inflammatory pathways exhibit a pivotal role in the reprogramming of WAT metabolism in physiological responses during cold, fasting and exercise. Thus, the underlying immunometabolic pathways may offer therapeutic targets to correct obesity and metabolic disease. Here, I review the current knowledge on the quantity and the quality of human adipose tissue macrophages (ATMΦ) and their impact on the bioenergetics of human adipocytes. The effects of ATMΦ and their secreted factors on mitochondrial function of white adipocytes are discussed, including recent research on MΦ as part of an immune signaling cascade involved in the 'browning' of WAT, which is defined as the conversion from white, energy-storing adipocytes into brown, energy-dissipating adipocytes.
Collapse
Affiliation(s)
- Michaela Keuper
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
89
|
Frey T, Swade K, Zwecker L, Llewellyn T, Vogt E, Monteferante K, English H. Monocyte Production of IFN-γ Is Interleukin-12 Dependent in a Model of Mevalonate Kinase Deficiency. J Interferon Cytokine Res 2019; 39:364-374. [PMID: 31013450 DOI: 10.1089/jir.2018.0126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mevalonate Kinase Deficiency (MKD) is an autoinflammatory disease caused by mutations in the mevalonate kinase gene, which produces an enzyme responsible for the production of isoprenoids in the mevalonate pathway. Patient data indicate that MKD is a multicytokine disease with increased plasma levels of cytokines such as tumor necrosis factor-α, interleukin (IL)-6, and interferon (IFN)-γ. To study the mechanisms responsible for these changes, the mevalonate pathway was inhibited with lovastatin in peripheral blood mononuclear cells (PBMCs) and monocytes isolated from the blood of healthy donors followed by stimulation with lipopolysaccharide (LPS) to induce an inflammatory response. Lovastatin treatment resulted in increased levels of IL-6, IL-12p40, and IFN-γ mRNA in both PBMCs and monocytes following LPS stimulation compared with control cells. An IL-12 neutralizing antibody blocked the increased levels of IFN-γ mRNA following lovastatin treatment in PBMCs indicating that this effect is dependent on IL-12. Flow cytometry experiments indicated that monocytes, not lymphocytes or granulocytes, are the source of increased IFN-γ and that both classical and nonclassical/intermediate monocytes express IFN-γ. These results indicate that blocking IL-12 or IFN- γ may be therapeutic options for MKD patients.
Collapse
Affiliation(s)
- Tiffany Frey
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| | - Katelyn Swade
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| | - Lindsey Zwecker
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| | - Tyler Llewellyn
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| | - Eric Vogt
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| | - Kim Monteferante
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| | - Heather English
- Department of Biology, Dickinson College, Carlisle, Pennsylvania
| |
Collapse
|
90
|
Zhang X, Cao X, Dang M, Wang H, Chen B, Du F, Li H, Zeng X, Guo C. Soluble receptor for advanced glycation end-products enhanced the production of IFN-γ through the NF-κB pathway in macrophages recruited by ischemia/reperfusion. Int J Mol Med 2019; 43:2507-2515. [PMID: 30942429 DOI: 10.3892/ijmm.2019.4152] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/21/2019] [Indexed: 11/06/2022] Open
Abstract
The current study investigated the role of sRAGE in the production of IFN‑γ in macrophages with I/R treatment. The number of macrophages in myocardial tissues treated with I/R with or without sRAGE was determined via immunohistochemical staining. Proliferative activity of macrophages was analyzed by a 5‑BrdU incorporation assay. Differentiation of macrophages was detected via immunofluorescence staining of iNOS (M1 macrophage marker). IFN‑γ production, due to sRAGE stimulation, in Raw 264.7 macrophages and the NF‑κB signaling pathway were measured using western blotting. A ChIP assay was used to examine the interactions between NF‑κB and the promoter of IFN‑γ. The results showed that the number of macrophages in I/R‑treated myocardial tissues was increased following sRAGE infusion. Proliferation of macrophages was increased significantly in the presence of sRAGE; after I/R treatment, the cells preferred to differentiate into M1 macrophages. IFN‑γ expression in Raw 264.7 macrophages was suppressed by an NF‑κB inhibitor (Bay117082) but enhanced by sRAGE, with or without I/R treatment. Furthermore, sRAGE increased the phosphorylation of IκB, IKK and NF‑κB, as well as the translocation of NF‑κB into the nucleus of Raw 264.7 macrophages, with or without I/R treatment. ChIP results showed that sRAGE promoted NF‑κB binding to the promoter of IFN‑γ in Raw 264.7 macrophages. Therefore, the findings of the present study indicated that sRAGE protected the heart from I/R injuries, which might be mediated by promoting infiltration and the differentiation of macrophages into M1, which would then synthesize and secrete IFN‑γ through activating the NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Xiuling Zhang
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Xianxian Cao
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Mengqiu Dang
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, P.R. China
| | - Buxing Chen
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Fenghe Du
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Huihua Li
- Department of Cardiology, Institute of Cardiovascular Disease, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, P.R. China
| | - Caixia Guo
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
91
|
Santinelli L, Statzu M, Pierangeli A, Frasca F, Bressan A, Pinacchio C, Nonne C, Turriziani O, Antonelli G, d'Ettorre G, Scagnolari C. Increased expression of IL-32 correlates with IFN-γ, Th1 and Tc1 in virologically suppressed HIV-1-infected patients. Cytokine 2019; 120:273-281. [PMID: 30910260 DOI: 10.1016/j.cyto.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/07/2019] [Accepted: 01/27/2019] [Indexed: 02/06/2023]
Abstract
Following recent attention focused on IL-32 as an important component involved in the inflammatory cytokine network, we speculated that IL-32's action on IFN-γ and IFN-γ secreting T cell subsets may help sustain the immune activation and dysregulation found in patients with HIV-1 achieving viral suppression. To explore this hypothesis, transcript levels of IL-32 and IFN-γ were evaluated in PBMC from 139 virologically suppressed HIV-1-infected patients and from 63 healthy individuals by Real Time RT-PCR assays. IL-32 and IFN-γ mRNA levels were also analyzed in CD4+ T cells, CD14+ monocytes and lamina propria lymphocytes (LPL) of the gut district in a subgroup of HIV-1-infected subjects. IFN-γ secreting CD4+ (Th1) and CD8+ (Tc1) T cell subset frequencies were evaluated in LPL by multiparametric flow cytometry. Gene expression results revealed that IL-32 and IFN-γ levels in PBMC from HIV-1-positive patients were significantly elevated compared to those from healthy donors, correlated with each other and increased with patient age. Both IL-32 and IFN-γ genes were also more strongly expressed in CD4+ T cells than in CD14+ monocytes. By contrast, IL-32 levels in LPL were comparable to those measured in PBMC, while IFN-γ levels were higher in PBMC than those in LPL. Negative correlations were found between IL-32 levels and the frequencies of Th1 and Tc1 subsets in gut mucosa. Collectively, our results provide the first evidence that IL-32 levels remain elevated in treated HIV-1-infected patients and correlate with IFN-γ, Th1 and Tc1 subsets.
Collapse
Affiliation(s)
- Letizia Santinelli
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Maura Statzu
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Alessia Bressan
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy; Microbiology and Virology Unit, Sapienza University Hospital, Rome, Italy.
| | - Claudia Pinacchio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| | - Chiara Nonne
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Ombretta Turriziani
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy; Microbiology and Virology Unit, Sapienza University Hospital, Rome, Italy.
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
92
|
Reynolds CJ, Chong DLW, Li Y, Black SL, Cutler A, Webster Z, Manji J, Altmann DM, Boyton RJ. Bioluminescent Reporting of In Vivo IFN-γ Immune Responses during Infection and Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2019; 202:2502-2510. [PMID: 30814307 PMCID: PMC6452029 DOI: 10.4049/jimmunol.1801453] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/30/2019] [Indexed: 12/17/2022]
Abstract
IFN-γ is a key cytokine of innate and adaptive immunity. It is important to understand temporal changes in IFN-γ production and how these changes relate to the role of IFN-γ in diverse models of infectious and autoimmune disease, making the ability to monitor and track IFN-γ production in vivo of a substantial benefit. IFN-γ ELISPOTs have been a central methodology to measure T cell immunity for many years. In this study, we add the capacity to analyze IFN-γ responses with high sensitivity and specificity, longitudinally, in vitro and in vivo. This allows the refinement of experimental protocols because immunity can be tracked in real-time through a longitudinal approach. We have generated a novel murine IFN-γ reporter transgenic model that allows IFN-γ production to be visualized and quantified in vitro and in vivo as bioluminescence using an imaging system. At baseline, in the absence of an inflammatory stimulus, IFN-γ signal from lymphoid tissue is detectable in vivo. Reporter transgenics are used in this study to track the IFN-γ response to Pseudomonas aeruginosa infection in the lung over time in vivo. The longitudinal development of the adaptive T cell immunity following immunization with Ag is identified from day 7 in vivo. Finally, we show that we are able to use this reporter transgenic to follow the onset of autoimmune T cell activation after regulatory T cell depletion in an established model of systemic autoimmunity. This IFN-γ reporter transgenic, termed “Gammaglow,” offers a valuable new modality for tracking IFN-γ immunity, noninvasively and longitudinally over time.
Collapse
Affiliation(s)
- Catherine J Reynolds
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| | - Deborah L W Chong
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| | - Yihan Li
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| | - S Lucas Black
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| | - Amy Cutler
- Transgenics and Embryonic Stem Cell Facility, Medical Research Council London Institute of Medical Sciences, London W12 ONN, United Kingdom
| | - Zoe Webster
- Transgenics and Embryonic Stem Cell Facility, Medical Research Council London Institute of Medical Sciences, London W12 ONN, United Kingdom
| | - Jiten Manji
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| | - Daniel M Altmann
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| | - Rosemary J Boyton
- Lung Immunology Group, Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London W12 ONN, United Kingdom; and
| |
Collapse
|
93
|
Novel IFN-γ ELISpot reveals robust T cell responses elicited after influenza nucleoprotein DNA vaccination in New Zealand White rabbits. Vaccine 2019; 37:903-909. [PMID: 30661837 DOI: 10.1016/j.vaccine.2019.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 02/06/2023]
Abstract
The New Zealand White rabbit is a highly accessible animal model which is regularly employed in biomedical research. However, the paucity of rabbit-specific reagents available limits its use in certain fields. Specifically, the lack of a reliable T cell assay has limited its employment in immune prophylactic and therapeutic studies. To address this inadequacy, we have developed an ELISpot assay to detect cellular immune responses (IFN-γ production) after antigenic stimulation. We have applied this assay to model the T cell responses elicited by a DNA vaccine. Immunization with an influenza nucleoprotein (NP) DNA vaccine revealed strong antigen-specific T cell responses in the peripheral blood mononuclear cell population. We believe this is the first report of such an assay in rabbit species, and it will become a useful tool to monitor in vivo responses to vaccines and permit the wider adoption of this model to measure immunological responses.
Collapse
|
94
|
Aranda-Souza MÂ, Lorena VMBD, Correia MTDS, Pereira-Neves A, Figueiredo RCBQD. A C-type lectin from Bothrops leucurus snake venom forms amyloid-like aggregates in RPMI medium and are efficiently phagocytosed by peritoneal macrophages. Toxicon 2019; 157:93-100. [DOI: 10.1016/j.toxicon.2018.11.309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/17/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
|
95
|
Tsukamoto M, Imai K, Ishimoto T, Komohara Y, Yamashita YI, Nakagawa S, Umezaki N, Yamao T, Kitano Y, Miyata T, Arima K, Okabe H, Baba Y, Chikamoto A, Ishiko T, Hirota M, Baba H. PD-L1 expression enhancement by infiltrating macrophage-derived tumor necrosis factor-α leads to poor pancreatic cancer prognosis. Cancer Sci 2018; 110:310-320. [PMID: 30426611 PMCID: PMC6317925 DOI: 10.1111/cas.13874] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy using anti‐PD‐1/PD‐L1 antibodies for several types of cancer has received considerable attention in recent decades. However, the molecular mechanism underlying PD‐L1 expression in pancreatic ductal adenocarcinoma (PDAC) cells has not been clearly elucidated. We investigated the clinical significance and regulatory mechanism of PD‐L1 expression in PDAC cells. Among the various cytokines tested, tumor necrosis factor (TNF)‐α upregulated PD‐L1 expression in PDAC cells through NF‐κB signaling. The induction of PD‐L1 expression was also caused by co‐culture with activated macrophages, and the upregulation was inhibited by neutralization with anti‐TNF‐α antibody after co‐culture with activated macrophages. PD‐L1 expression in PDAC cells was positively correlated with macrophage infiltration in tumor stroma of human PDAC tissues. In addition, survival analysis revealed that high PD‐L1 expression was significantly associated with poor prognosis in 235 PDAC patients and especially in patients harboring high CD8‐positive T‐cell infiltration. These findings indicate that tumor‐infiltrating macrophage‐derived TNF‐α could be a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Masayo Tsukamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoki Umezaki
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takanobu Yamao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kitano
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsunori Miyata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kota Arima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirohisa Okabe
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Akira Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatoshi Ishiko
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masahiko Hirota
- Department of Surgery, Kumamoto Regional Medical Center, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
96
|
Gomes TS, Gjiknuri J, Magnet A, Vaccaro L, Ollero D, Izquierdo F, Fenoy S, Hurtado C, Del Águila C. The Influence of Acanthamoeba- Legionella Interaction in the Virulence of Two Different Legionella Species. Front Microbiol 2018; 9:2962. [PMID: 30568639 PMCID: PMC6290054 DOI: 10.3389/fmicb.2018.02962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/16/2018] [Indexed: 12/15/2022] Open
Abstract
The genus Legionella comprises more than 60 species, and about half are associated with infection. Legionella pneumophila is the most commonly associated with these infections and by far the most studied, but L. non-pneumophila species, such as L. feeleii, L. anisa, etc., may also present clinical importance. Free-living amoebae are their preferred environmental host, where these bacteria not only survive but also succeed in multiplying, and this relationship can lead to an increase in bacterial virulence. The goal of this study was to evaluate the alterations of Legionella pathogenicity due to its interaction with Acanthamoeba. For this, the expression of protein effectors SdhA, LegK2, and SidK were evaluated in L. pneumophila and L. feeleii, before and after infecting Acanthamoeba. Additionally, the host response was evaluated by measuring the production of IL-6, IL-8, and IFN-γ in infected macrophages. Regarding the virulence factors, an increase in SdhA expression was observed after these bacteria infected Acanthamoeba, with a higher increase in the macrophage cultures infected with L. feeleii. Also, an increase in the expression of LegK2 was observed after infecting Acanthamoeba, but it was more intense in the cultures infected with L. pneumophila. With regard to SidK, it was increased in L. feeleii after infecting Acanthamoeba, however the same effect was not observed for L. pneumophila. In cytokine production, the effect on IL-6 and IL-8 was similar for both cytokines, increasing their concentration, but higher production was observed in the cultures infected with L. feeleii, even though it demonstrated slightly lower production with the inoculum obtained from Acanthamoeba. Concerning IFN-γ, induction was observed in both species but higher in the infection by L. pneumophila. Nevertheless, it is not known if this induction is enough to promote an efficient immune response against either L. pneumophila or L. feeleii. Altogether, these alterations seem to increase L. feeleii virulence after infecting Acanthamoeba. However, this increase does not seem to turn L. feeleii as virulent as L. pneumophila. More studies are necessary to understand the aspects influenced in these bacteria by their interaction with Acanthamoeba and, thus, identify targets to be used in future therapeutic approaches.
Collapse
Affiliation(s)
- Thiago Santos Gomes
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain.,CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Julia Gjiknuri
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Angela Magnet
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Lucianna Vaccaro
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Dolores Ollero
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Fernando Izquierdo
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Soledad Fenoy
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Carolina Hurtado
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Carmen Del Águila
- Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
97
|
Atkins C, Miao J, Kalveram B, Juelich T, Smith JK, Perez D, Zhang L, Westover JLB, Van Wettere AJ, Gowen BB, Wang Z, Freiberg AN. Natural History and Pathogenesis of Wild-Type Marburg Virus Infection in STAT2 Knockout Hamsters. J Infect Dis 2018; 218:S438-S447. [PMID: 30192975 PMCID: PMC6249581 DOI: 10.1093/infdis/jiy457] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Marburg virus (MARV; family Filoviridae) causes sporadic outbreaks of Marburg hemorrhagic fever in sub-Saharan Africa with case fatality rates reaching 90%. Wild-type filoviruses, including MARV and the closely related Ebola virus, are unable to suppress the type I interferon response in rodents, and therefore require adaptation of the viruses to cause disease in immunocompetent animals. In the current study, we demonstrate that STAT2 knockout Syrian hamsters are susceptible to infection with different wild-type MARV variants. MARV Musoke causes a robust and systemic infection resulting in lethal disease. Histopathological findings share features similar to those observed in human patients and other animal models of filovirus infection. Reverse-transcription polymerase chain reaction analysis of host transcripts shows a dysregulation of the innate immune response. Our results demonstrate that the STAT2 knockout hamster represents a novel small animal model of severe MARV infection and disease without the requirement for virus adaptation.
Collapse
Affiliation(s)
- Colm Atkins
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Jinxin Miao
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Birte Kalveram
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Terry Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Jennifer K Smith
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - David Perez
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Lihong Zhang
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Jonna L B Westover
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Arnaud J Van Wettere
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Brian B Gowen
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston
| |
Collapse
|
98
|
Abstract
Interferon-gamma (IFNG) has long been implicated as a central orchestrator of antitumor immune responses in the elimination stage of the immunoediting paradigm. However, mounting evidence suggests that IFNG may also have important and significant protumor roles to play in the equilibrium and escape phases through its regulatory effects on immunoevasive functions that promote tumorigenesis. These seemingly contradictory effects of IFNG undoubtedly play profound roles in not only the activation of inflammatory response to cancer but also in the determination of its outcome. In the face of the recent explosion of anticancer immunotherapeutic strategies in the clinic, it is critical that a complete understanding is achieved of the underpinnings of the mechanisms that determine the two faces of IFNG signaling in cancer. Here, the current state of this dichotomy is reviewed.
Collapse
Affiliation(s)
- M Raza Zaidi
- Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
99
|
Impact of the GK-1 adjuvant on peritoneal macrophages gene expression and phagocytosis. Immunol Lett 2018; 201:20-30. [DOI: 10.1016/j.imlet.2018.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/10/2018] [Accepted: 10/18/2018] [Indexed: 11/23/2022]
|
100
|
Lawrence S, Ismail F, Jamal SZ, Whalen MM. Tributyltin stimulates synthesis of interferon gamma and tumor necrosis factor alpha in human lymphocytes. J Appl Toxicol 2018; 38:1081-1090. [PMID: 29532501 PMCID: PMC5997500 DOI: 10.1002/jat.3617] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Abstract
Tributyltin (TBT) is found in human blood and other tissues and thus is of considerable concern as to its effects on human health. Previous studies have demonstrated that TBT has detrimental effects on immune function. Recently, we found that exposures to TBT caused increased secretion of two important proinflammatory cytokines, tumor necrosis factor alpha (TNFα) and interferon gamma (IFNγ). Elevation of either of these cytokines has the potential to cause chronic inflammation, which is an important factor in a number of diseases including cancer. The current study examined the mechanism of TBT-induced elevations of TNFα and IFNγ secretion and found that the p38 mitogen-activated protein kinase pathway was essential to the ability of TBT to stimulate secretion. Additionally, this study demonstrated that increased secretion of these cytokines was due to TBT-induced increases in their overall synthesis, rather than simply being due to an increase in the release of already formed proteins. The TBT-induced increases in synthesis were evident within 6 hours of exposure. The p38 mitogen-activated protein kinase pathway is also necessary for the TBT-induced increases in both TNFα and IFNγ synthesis. The role of increased transcription of TNFα and IFNγ mRNA in response to TBT exposures as a possible explanation for the increased synthesis of these cytokines was also examined. It was found that increased mRNA levels did not appear to explain fully the increases in either TNFα or IFNγ synthesis. Thus, TBT is able to increase secretion of two important proinflammatory cytokines by increasing their synthesis.
Collapse
Affiliation(s)
- Shanieek Lawrence
- Departments of Biological Sciences, Tennessee State University, Nashville, TN, 37209, USA
| | - Farah Ismail
- Departments of Chemistry, Tennessee State University, Nashville, TN, 37209, USA
| | - Sarah Z Jamal
- Departments of Chemistry, Tennessee State University, Nashville, TN, 37209, USA
| | - Margaret M Whalen
- Departments of Chemistry, Tennessee State University, Nashville, TN, 37209, USA
| |
Collapse
|