1
|
Solomon A, Bossel Ben-Moshe N, Hoffman D, Trzebanski S, Yehezkel D, Vainman L, Netea MG, Avraham R. Early and delayed STAT1-dependent responses drive local trained immunity of macrophages in the spleen. eLife 2025; 13:RP100922. [PMID: 39819562 PMCID: PMC11737870 DOI: 10.7554/elife.100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
Trained immunity (TI) is the process wherein innate immune cells gain functional memory upon exposure to specific ligands or pathogens, leading to augmented inflammatory responses and pathogen clearance upon secondary exposure. While the differentiation of hematopoietic stem cells (HSCs) and reprogramming of bone marrow (BM) progenitors are well-established mechanisms underpinning durable TI protection, remodeling of the cellular architecture within the tissue during TI remains underexplored. Here, we study the effects of peritoneal Bacillus Calmette-Guérin (BCG) administration to find TI-mediated protection in the spleen against a subsequent heterologous infection by the Gram-negative pathogen Salmonella Typhimurium (S.Tm). Utilizing single cell RNA-sequencing and flow cytometry, we discerned STAT1-regulated genes in TI-associated resident and recruited splenic myeloid populations. The temporal dynamics of TI were further elucidated, revealing both early and delayed myeloid subsets with time-dependent, cell-type-specific STAT1 signatures. Using lineage tracing, we find that tissue-resident red pulp macrophages (RPM), initially depleted by BCG exposure, are restored from both tissue-trained, self-renewing macrophages and from bone marrow-derived progenitors, fostering long lasting local defense. Early inhibition of STAT1 activation, using specific JAK-STAT inhibitors, reduces both RPM loss and recruitment of trained monocytes. Our study suggests a temporal window soon after BCG vaccination, in which STAT1-dependent activation of long-lived resident cells in the tissue mediates localized protection.
Collapse
Affiliation(s)
- Aryeh Solomon
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Noa Bossel Ben-Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Dotan Hoffman
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Sébastien Trzebanski
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Dror Yehezkel
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Leia Vainman
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical CenterNijmegenNetherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of BonnBonnGermany
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
2
|
Jaime-Sánchez E, Lara-Ramírez EE, López-Ramos JE, Ramos-González EJ, Cisneros-Méndez AL, Oropeza-Valdez JJ, Zenteno-Cuevas R, Martínez-Aguilar G, Bastian Y, Castañeda-Delgado JE, Serrano CJ, Enciso-Moreno JA. Potential molecular patterns for tuberculosis susceptibility in diabetic patients with poor glycaemic control: a pilot study. Mol Genet Genomics 2024; 299:60. [PMID: 38801463 DOI: 10.1007/s00438-024-02139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 04/06/2024] [Indexed: 05/29/2024]
Abstract
Type 2 diabetes (DM2) is an increasingly prevalent disease that challenges tuberculosis (TB) control strategies worldwide. It is significant that DM2 patients with poor glycemic control (PDM2) are prone to developing tuberculosis. Furthermore, elucidating the molecular mechanisms that govern this susceptibility is imperative to address this problem. Therefore, a pilot transcriptomic study was performed. Human blood samples from healthy controls (CTRL, HbA1c < 6.5%), tuberculosis (TB), comorbidity TB-DM2, DM2 (HbA1c 6.5-8.9%), and PDM2 (HbA1c > 10%) groups (n = 4 each) were analyzed by differential expression using microarrays. We use a network strategy to identify potential molecular patterns linking the differentially expressed genes (DEGs) specific for TB-DM2 and PDM2 (p-value < 0.05, fold change > 2). We define OSM, PRKCD, and SOCS3 as key regulatory genes (KRGs) that modulate the immune system and related pathways. RT-qPCR assays confirmed upregulation of OSM, PRKCD, and SOCS3 genes (p < 0.05) in TB-DM2 patients (n = 18) compared to CTRL, DM2, PDM2, or TB groups (n = 17, 19, 15, and 9, respectively). Furthermore, OSM, PRKCD, and SOCS3 were associated with PDM2 susceptibility pathways toward TB-DM2 and formed a putative protein-protein interaction confirmed in STRING. Our results reveal potential molecular patterns where OSM, PRKCD, and SOCS3 are KRGs underlying the compromised immune response and susceptibility of patients with PDM2 to develop tuberculosis. Therefore, this work paved the way for fundamental research of new molecular targets in TB-DM2. Addressing their cellular implications, and the impact on the diagnosis, treatment, and clinical management of TB-DM2 could help improve the strategy to end tuberculosis for this vulnerable population.
Collapse
Affiliation(s)
- Elena Jaime-Sánchez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, Tamaulipas, México
- Área de Ciencias de La Salud, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara, Zacatecas, México
- Unidad de Investigación Biomédica de Zacatecas, IMSS, Zacatecas, México
| | - Edgar E Lara-Ramírez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, Tamaulipas, México
- Unidad de Investigación Biomédica de Zacatecas, IMSS, Zacatecas, México
| | - Juan Ernesto López-Ramos
- Academia de Ciencias Químico-Biológicas, Instituto Politécnico Nacional, Centro de Estudios Científicos y Tecnológicos No. 18, Zacatecas, México
| | | | | | - Juan José Oropeza-Valdez
- Human Systems Biology Laboratory. Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | | | | | - Yadira Bastian
- Instituto de Física, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Julio Enrique Castañeda-Delgado
- Unidad de Investigación Biomédica de Zacatecas, IMSS, Zacatecas, México
- Investigador por Mexico/Catedras CONAHCYT, Consejo nacional de Humanidades, Ciencias y Tecnologias, Ciudad de Mexico, México
- Consejo Nacional de Ciencia y Tecnologia, CONACYT, Ciudad de Mexico, México
| | | | - José Antonio Enciso-Moreno
- Unidad de Investigación Biomédica de Zacatecas, IMSS, Zacatecas, México.
- Facultad de Química, Cerro de Las Campanas S/N, Universidad Autónoma de Querétaro, Colonia Las Campanas, Centro Universitario, C.P. 76010, Querétaro, México.
| |
Collapse
|
3
|
Ma Q, Chen J, Kong X, Zeng Y, Chen Z, Liu H, Liu L, Lu S, Wang X. Interactions between CNS and immune cells in tuberculous meningitis. Front Immunol 2024; 15:1326859. [PMID: 38361935 PMCID: PMC10867975 DOI: 10.3389/fimmu.2024.1326859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
The central nervous system (CNS) harbors its own special immune system composed of microglia in the parenchyma, CNS-associated macrophages (CAMs), dendritic cells, monocytes, and the barrier systems within the brain. Recently, advances in the immune cells in the CNS provided new insights to understand the development of tuberculous meningitis (TBM), which is the predominant form of Mycobacterium tuberculosis (M.tb) infection in the CNS and accompanied with high mortality and disability. The development of the CNS requires the protection of immune cells, including macrophages and microglia, during embryogenesis to ensure the accurate development of the CNS and immune response following pathogenic invasion. In this review, we summarize the current understanding on the CNS immune cells during the initiation and development of the TBM. We also explore the interactions of immune cells with the CNS in TBM. In the future, the combination of modern techniques should be applied to explore the role of immune cells of CNS in TBM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuihua Lu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Xiaomin Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Novita BD, Tjahjono Y, Wijaya S, Theodora I, Erwin F, Halim SW, Hendrawan B, Jaya DK, Tahalele PL. Characterization of chemokine and cytokine expression pattern in tuberculous lymphadenitis patient. Front Immunol 2022; 13:983269. [PMID: 36439164 PMCID: PMC9692123 DOI: 10.3389/fimmu.2022.983269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/20/2022] [Indexed: 08/30/2023] Open
Abstract
INTRODUCTION C-C chemokine receptor-2 (CCR-2) and C-C chemokine ligand-5 (CCL-5) play an important role in the migration of monocytes, macrophages, dendritic cells, and activated T cells against Mycobacterium tuberculosis (M.tb). Meanwhile, signal transducer and activator of transcription 3 (STAT-3) and suppressor of cytokine signaling 3 (SOCS-3), activated by interleukin (IL)-6 and IL-10 in tuberculosis (TB) infection, play an important role in phagocytosis, inflammation, and granulomatous-forming processes that may lead to TB treatment success or failure. However, there are no data about the expression of those markers in tuberculous lymphadenitis. The characterization of those markers is very critical to put a fundamental basis to understand the homing mechanism of tuberculous lymphadenitis. AIM OF STUDY The specific objective of this study is to characterize the expression pattern of CCR-2-CCL-5, IL-6, IL-10, STAT-3, and SOCS-3 in tuberculous lymphadenitis. METHODS The study was performed on 27 cases of tuberculous lymphadenitis node biopsies. The diagnosis of tuberculous lymphadenitis was based on the clinical criteria and the presence of the histological feature characteristic of TB granulomas. Afterward, immunohistochemistry was stained with CCR-2, CCL-5, IL-6, IL-10, STAT-3, and SOCS-3. A semiquantitative analysis of IHC images was performed to examine protein expression in stained preparations. The expression was also manually counted. RESULTS Compared with the normal area, both lymphocytes and macrophages expressed strongly CCR-2-, CCL-5, and IL-6, while IL-10, STAT-3-, and SOCS-3- were expressed lowly. There was a strong positive correlation between CCR-2 with IL-6 (p = 0,83) and IL-10 (p = 0,83). CONCLUSION The chronic infection process of tuberculous lymphadenitis was characterized by the expression of IL-10low, STAT-3low, SOCS-3low, CCR-2high, CCL-5high, and IL-6high. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov, identifier NCT05202548.
Collapse
Affiliation(s)
| | - Yudy Tjahjono
- Faculty of Pharmacy, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - Sumi Wijaya
- Faculty of Pharmacy, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - Imelda Theodora
- Faculty of Medicine, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - Ferdinand Erwin
- Faculty of Medicine, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - Stefan Wilson Halim
- Faculty of Medicine, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - Bobby Hendrawan
- Faculty of Medicine, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - David Karunia Jaya
- Faculty of Medicine, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| | - Paul L. Tahalele
- Faculty of Medicine, Widya Mandala Surabaya Catholic University, Surabaya, Indonesia
| |
Collapse
|
5
|
Magoulopoulou A, Qian X, Pediatama Setiabudiawan T, Marco Salas S, Yokota C, Rottenberg ME, Nilsson M, Carow B. Spatial Resolution of Mycobacterium tuberculosis Bacteria and Their Surrounding Immune Environments Based on Selected Key Transcripts in Mouse Lungs. Front Immunol 2022; 13:876321. [PMID: 35663950 PMCID: PMC9157500 DOI: 10.3389/fimmu.2022.876321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) bacilli are the causative agent of tuberculosis (TB), a major killer of mankind. Although it is widely accepted that local interactions between Mtb and the immune system in the tuberculous granuloma determine whether the outcome of infection is controlled or disseminated, these have been poorly studied due to methodological constraints. We have recently used a spatial transcriptomic technique, in situ sequencing (ISS), to define the spatial distribution of immune transcripts in TB mouse lungs. To further contribute to the understanding of the immune microenvironments of Mtb and their local diversity, we here present two complementary automated bacteria-guided analysis pipelines. These position 33 ISS-identified immune transcripts in relation to single bacteria and bacteria clusters. The analysis was applied on new ISS data from lung sections of Mtb-infected C57BL/6 and C3HeB/FeJ mice. In lungs from C57BL/6 mice early and late post infection, transcripts that define inflammatory macrophages were enriched at subcellular distances to bacteria, indicating the activation of infected macrophages. In contrast, expression patterns associated to antigen presentation were enriched in non-infected cells at 12 weeks post infection. T-cell transcripts were evenly distributed in the tissue. In Mtb-infected C3HeB/FeJ mice, transcripts characterizing activated macrophages localized in apposition to small bacteria clusters, but not in organized granulomas. Despite differences in the susceptibility to Mtb, the transcript patterns found around small bacteria clusters of C3HeB/FeJ and C57BL/6 mice were similar. Altogether, the presented tools allow us to characterize in depth the immune cell populations and their activation that interact with Mtb in the infected lung.
Collapse
Affiliation(s)
- Anastasia Magoulopoulou
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Xiaoyan Qian
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Todia Pediatama Setiabudiawan
- Department of Microbiology, Tumor and Cell Biology and Centre for Tuberculosis Research, Karolinska Institutet, Solna, Sweden
| | - Sergio Marco Salas
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Chika Yokota
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology and Centre for Tuberculosis Research, Karolinska Institutet, Solna, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Berit Carow
- Department of Microbiology, Tumor and Cell Biology and Centre for Tuberculosis Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
6
|
Sobah ML, Liongue C, Ward AC. SOCS Proteins in Immunity, Inflammatory Diseases, and Immune-Related Cancer. Front Med (Lausanne) 2021; 8:727987. [PMID: 34604264 PMCID: PMC8481645 DOI: 10.3389/fmed.2021.727987] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/16/2021] [Indexed: 01/10/2023] Open
Abstract
Cytokine signaling represents one of the cornerstones of the immune system, mediating the complex responses required to facilitate appropriate immune cell development and function that supports robust immunity. It is crucial that these signals be tightly regulated, with dysregulation underpinning immune defects, including excessive inflammation, as well as contributing to various immune-related malignancies. A specialized family of proteins called suppressors of cytokine signaling (SOCS) participate in negative feedback regulation of cytokine signaling, ensuring it is appropriately restrained. The eight SOCS proteins identified regulate cytokine and other signaling pathways in unique ways. SOCS1–3 and CISH are most closely involved in the regulation of immune-related signaling, influencing processes such polarization of lymphocytes and the activation of myeloid cells by controlling signaling downstream of essential cytokines such as IL-4, IL-6, and IFN-γ. SOCS protein perturbation disrupts these processes resulting in the development of inflammatory and autoimmune conditions as well as malignancies. As a consequence, SOCS proteins are garnering increased interest as a unique avenue to treat these disorders.
Collapse
Affiliation(s)
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Institue of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Institue of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
7
|
Beyranvand Nejad E, Labrie C, van Elsas MJ, Kleinovink JW, Mittrücker HW, Franken KLMC, Heink S, Korn T, Arens R, van Hall T, van der Burg SH. IL-6 signaling in macrophages is required for immunotherapy-driven regression of tumors. J Immunother Cancer 2021; 9:e002460. [PMID: 33879600 PMCID: PMC8061866 DOI: 10.1136/jitc-2021-002460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND High serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective. METHODS IL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1. CONTROL Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6rafl/fl×LysMcre+ mice. RESULTS Our therapeutic vaccination protocol elicits a strong tumor-specific CD8+ T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8+ T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6rafl/fl×LysMcre+ but not cre-negative control mice, while no differences in the vaccine-induced CD8+ T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages. CONCLUSION IL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic/drug effects
- Female
- Injections, Subcutaneous
- Interleukin-6/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Papillomavirus E7 Proteins/administration & dosage
- Papillomavirus E7 Proteins/immunology
- Phenotype
- Receptors, Interleukin-6/genetics
- Receptors, Interleukin-6/metabolism
- Signal Transduction
- Tumor Burden/drug effects
- Tumor-Associated Macrophages/drug effects
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
- Mice
Collapse
Affiliation(s)
- Elham Beyranvand Nejad
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Camilla Labrie
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Marit J van Elsas
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Jan Willem Kleinovink
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kees L M C Franken
- Department of Immunology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sylvia Heink
- Experimental Neuroimmunology, Technische Universität München, Munchen, Bayern, Germany
| | - Thomas Korn
- Experimental Neuroimmunology, Technische Universität München, Munchen, Bayern, Germany
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Thorbald van Hall
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sjoerd H van der Burg
- Medical Oncology, Oncode institute, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
8
|
Ritter K, Rousseau J, Hölscher C. The Role of gp130 Cytokines in Tuberculosis. Cells 2020; 9:E2695. [PMID: 33334075 PMCID: PMC7765486 DOI: 10.3390/cells9122695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Protective immune responses to Mycobacterium tuberculosis (Mtb) infection substantially depend on a delicate balance within cytokine networks. Thus, immunosuppressive therapy by cytokine blockers, as successfully used in the management of various chronic inflammatory diseases, is often connected with an increased risk for tuberculosis (TB) reactivation. Hence, identification of alternative therapeutics which allow the treatment of inflammatory diseases without compromising anti-mycobacterial immunity remains an important issue. On the other hand, in the context of novel therapeutic approaches for the management of TB, host-directed adjunct therapies, which combine administration of antibiotics with immunomodulatory drugs, play an increasingly important role, particularly to reduce the duration of treatment. In both respects, cytokines/cytokine receptors related to the common receptor subunit gp130 may serve as promising target candidates. Within the gp130 cytokine family, interleukin (IL)-6, IL-11 and IL-27 are most explored in the context of TB. This review summarizes the differential roles of these cytokines in protection and immunopathology during Mtb infection and discusses potential therapeutic implementations with respect to the aforementioned approaches.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
| | - Jasmin Rousseau
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
| | - Christoph Hölscher
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, D-23845 Borstel, Germany
| |
Collapse
|
9
|
Ji XC, Shi YJ, Zhang Y, Chang MZ, Zhao G. Reducing Suppressors of Cytokine Signaling-3 (SOCS3) Expression Promotes M2 Macrophage Polarization and Functional Recovery After Intracerebral Hemorrhage. Front Neurol 2020; 11:586905. [PMID: 33281724 PMCID: PMC7688919 DOI: 10.3389/fneur.2020.586905] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/02/2020] [Indexed: 12/24/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a fatal subtype of stroke, and effective interventions to improve the functional outcomes are still lacking. Suppressor of cytokine signaling 3 (SOCS3) plays critical roles in the inflammatory response by negatively regulating cytokine-Jak-Stat signaling. However, the role of SOCS3 in the regulation of macrophage polarization is highly controversial and the fine regulation exerted by SOCS3 needs further understanding. In this study, rat ICH models were established by infusion of collagenase into the caudate nucleus. To decrease SOCS3 expression into microglia/macrophages in the hemorrhagic lesion area, we injected lentiviral short hairpin RNA (shSOCS3) (Lenti-shSOCS3) into the hematoma cavity at 24 h following ICH. We found that the number of iNOS-positive cells (M1 phenotype) was significantly reduced, whereas arginase-1-positive cells (M2 phenotype) were markedly elevated in animals that received Lenti-shSOCS3 injections compared with those in the Lenti-EGFP and saline groups. The increase in arginase-1-positive cells was associated with a significantly lower pro-inflammatory microenvironment, which included the downregulation of pro-inflammatory cytokines [interleukin (IL)-1β, IL-6, and TNF-α] and concurrent upregulation of anti-inflammatory (IL-10) mediators. In addition, this marked shift toward the M2 phenotype was associated with suppressed NF-κB activation. Furthermore, these changes notably enhanced the neuroprotective effects and functional recovery in Lenti-shSOCS3-injected animals. Our findings indicated that reduction in SOCS3 expression caused a marked bias toward the M2 phenotype and ameliorated the inflammatory microenvironment, which enhanced neuroprotective effects and resulted in notable improvement in functional recovery after ICH.
Collapse
Affiliation(s)
- Xin-Chao Ji
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China.,Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ya-Jun Shi
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Zhang
- Affiliated Bayi Brain Hospital, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Ming-Ze Chang
- Department of Neurology, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
10
|
Hombrebueno JR, Lynch A, Byrne EM, Obasanmi G, Kissenpfennig A, Chen M, Xu H. Hyaloid Vasculature as a Major Source of STAT3 + (Signal Transducer and Activator of Transcription 3) Myeloid Cells for Pathogenic Retinal Neovascularization in Oxygen-Induced Retinopathy. Arterioscler Thromb Vasc Biol 2020; 40:e367-e379. [PMID: 33115265 DOI: 10.1161/atvbaha.120.314567] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Myeloid cells are critically involved in inflammation-induced angiogenesis, although their pathogenic role in the ischemic retina remains controversial. We hypothesize that myeloid cells contribute to pathogenic neovascularization in retinopathy of prematurity through STAT3 (signal transducer and activator of transcription 3) activation. Approach and Results: Using the mouse model of oxygen-induced retinopathy, we show that myeloid cells (CD45+IsolectinB4 [IB4]+) and particularly M2-type macrophages (CD45+ Arg1+), comprise a major source of STAT3 activation (pSTAT3) in the immature ischemic retina. Most of the pSTAT3-expressing myeloid cells concentrated at the hyaloid vasculature and their numbers were strongly correlated with the severity of pathogenic neovascular tuft formation. Pharmacological inhibition of STAT3 reduced the load of IB4+ cells in the hyaloid vasculature and significantly reduced the formation of pathogenic neovascular tufts during oxygen-induced retinopathy, leading to improved long-term visual outcomes (ie, increased retinal thickness and scotopic b-wave electroretinogram responses). Genetic deletion of SOCS3 (suppressor of cytokine signaling 3), an endogenous inhibitor of STAT3, in myeloid cells, enhanced pathological and physiological neovascularization in oxygen-induced retinopathy, indicating that myeloid-STAT3 signaling is crucial for retinal angiogenesis. CONCLUSIONS Circulating myeloid cells may migrate to the immature ischemic retina through the hyaloid vasculature and contribute to retinal neovascularization via activation of STAT3. Understanding how STAT3 modulates myeloid cells for vascular repair/pathology may provide novel therapeutic options in pathogenic angiogenesis.
Collapse
Affiliation(s)
- Jose R Hombrebueno
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.).,Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (J.R.H.)
| | - Aisling Lynch
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Eimear M Byrne
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Gideon Obasanmi
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Adrien Kissenpfennig
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Mei Chen
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Heping Xu
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| |
Collapse
|
11
|
Cevey ÁC, Penas FN, Alba Soto CD, Mirkin GA, Goren NB. IL-10/STAT3/SOCS3 Axis Is Involved in the Anti-inflammatory Effect of Benznidazole. Front Immunol 2019; 10:1267. [PMID: 31214200 PMCID: PMC6558013 DOI: 10.3389/fimmu.2019.01267] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Anti-parasitic treatment for Chagas disease mainly relies on benznidazole, which is virtually the only drug available in the market. Besides its anti-parasitic effects, benznidazole has anti-inflammatory properties. In this work we studied the mechanisms involved in the latter, demonstrating the participation of the IL-10/STAT3/SOCS3 pathway. To achieve this goal, the anti-inflammatory properties of benznidazole were studied using an in vitro model of cardiomyocyte primary culture stimulated with LPS. LPS increased both SOCS3 expression and STAT3 phosphorylation. The addition of benznidazole increased their expression even further. Specific inhibition of STAT3 precluded this effect, suggesting a role for STAT3 in the increase of SOCS3 expression induced by benznidazole. To assess the participation of SOCS3 in the anti-inflammatory effect of benznidazole, we accomplished specific knockdown of SOCS3 with siRNA. Silencing of SOCS3 in cardiomyocytes precluded the inhibitory effects of benznidazole on TNF-α, IL-6, iNOS expression and NO release. Moreover, in the absence of SOCS3, benznidazole could neither prevent IKK phosphorylation nor IκBα degradation, supporting the notion that SOCS3 is required for the benznidazole-mediated inhibition of the NF-κB pathway. Previously, we demonstrated that IL-10 increases the expression of SOCS3 in cultured cardiomyocytes. Here, we found that benznidazole shows a trend to increased IL-10 expression. To evaluate whether benznidazole increased SOCS3 in an IL-10-dependent manner, cardiomyocytes from IL-10 knockout mice were pre-treated with benznidazole and stimulated with LPS. Benznidazole neither inhibited NO release nor avoid IKK phosphorylation or IκBα degradation, showing that IL-10 is required for benznidazole-mediated inhibition of NF-κB. Moreover, exogenous addition of IL-10 to IL-10 knockout cardiomyocytes restored the inhibitory effect of benznidazole on NO release. The results reported herein show, for the first time, that the IL-10/STAT3/SOCS3 axis is involved in the anti-inflammatory effects of benznidazole. These findings may add up to new therapeutic strategies for chronic Chagas disease given its inflammatory nature.
Collapse
Affiliation(s)
- Ágata C Cevey
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico N Penas
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Catalina D Alba Soto
- Facultad de Medicina, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gerardo A Mirkin
- Facultad de Medicina, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora B Goren
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
12
|
Harling K, Adankwah E, Güler A, Afum-Adjei Awuah A, Adu-Amoah L, Mayatepek E, Owusu-Dabo E, Nausch N, Jacobsen M. Constitutive STAT3 phosphorylation and IL-6/IL-10 co-expression are associated with impaired T-cell function in tuberculosis patients. Cell Mol Immunol 2019; 16:275-287. [PMID: 30886421 PMCID: PMC6460487 DOI: 10.1038/cmi.2018.5] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/27/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023] Open
Abstract
T-cells critically contribute to protection against Mycobacterium tuberculosis infection, and impaired T-cell responses can lead to disease progression. Pro-inflammatory and immunosuppressive cytokines affect T-cells, and fine-tuned regulation of cytokine signaling via the Jak/STAT signaling pathways is crucial for appropriate T-cell function. Constitutive STAT3 phosphorylation as a consequence of aberrant cytokine signaling has been described to occur in pathognomonic T-cell responses in inflammatory and autoimmune diseases. We characterized blood samples from tuberculosis patients (n=28) and healthy contacts (n=28) from Ghana for M. tuberculosis-specific T-cell responses, constitutive cytokine production, and SOCS3 and pSTAT3 expression. Lentiviral modulation of primary CD4+ T-cells was performed to determine the effects of SOCS3 on T-cell functions. T-cells from tuberculosis patients expressed higher levels of IL-10 and IL-6 and lower levels of T helper type (TH)17 cytokines after M. tuberculosis-specific stimulation compared to healthy contacts. In addition, tuberculosis patients had higher IL-10 and IL-6 levels in the supernatants of non-stimulated immune cells and plasma samples compared to healthy contacts. Notably, aberrant cytokine expression was accompanied by high constitutive pSTAT3 levels and SOCS3 expression in T-cells. Multivariate analysis identified an IL-6/IL-10 co-expression-based principal component in tuberculosis patients that correlated with high pSTAT3 levels. SOCS3 contributed to a regulatory component, and tuberculosis patients with high SOCS3 expression showed decreased TH1 cytokine expression and impaired IL-2-induced STAT5 phosphorylation. SOCS3 over-expression in primary CD4+ T-cells confirmed the SOCS3 inhibitory function on IL-2-induced STAT5 phosphorylation. We conclude that constitutive pSTAT3 and high SOCS3 expression are influential factors that indicate impaired T-cell functions in tuberculosis patients.
Collapse
Affiliation(s)
- Kirstin Harling
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, 40225, Duesseldorf, Germany
| | - Ernest Adankwah
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, 40225, Duesseldorf, Germany
| | - Alptekin Güler
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, 40225, Duesseldorf, Germany
| | - Anthony Afum-Adjei Awuah
- Kumasi Centre for collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
- School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Louis Adu-Amoah
- Kumasi Centre for collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, 40225, Duesseldorf, Germany
| | - Ellis Owusu-Dabo
- Kumasi Centre for collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
- School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Norman Nausch
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, 40225, Duesseldorf, Germany
| | - Marc Jacobsen
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, 40225, Duesseldorf, Germany.
| |
Collapse
|
13
|
Gao Y, Zhao H, Wang P, Wang J, Zou L. The roles of SOCS3 and STAT3 in bacterial infection and inflammatory diseases. Scand J Immunol 2018; 88:e12727. [PMID: 30341772 DOI: 10.1111/sji.12727] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/11/2018] [Accepted: 10/13/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Yu Gao
- Translational Neuroscience & Neural Regeneration and Repair Institute/Institute of Cell Therapy; The People's Hospital of China Three Gorges University; Yichang China
- Department of Microbiology, Tumor and Cell Biology; Karolinska Institutet; Stockholm Sweden
| | - Honglei Zhao
- Translational Neuroscience & Neural Regeneration and Repair Institute/Institute of Cell Therapy; The People's Hospital of China Three Gorges University; Yichang China
- Department of Oncology-Pathology; Karolinska Institutet; Stockholm Sweden
| | - Peng Wang
- Translational Neuroscience & Neural Regeneration and Repair Institute/Institute of Cell Therapy; The People's Hospital of China Three Gorges University; Yichang China
| | - Jun Wang
- Translational Neuroscience & Neural Regeneration and Repair Institute/Institute of Cell Therapy; The People's Hospital of China Three Gorges University; Yichang China
| | - Lili Zou
- Translational Neuroscience & Neural Regeneration and Repair Institute/Institute of Cell Therapy; The People's Hospital of China Three Gorges University; Yichang China
| |
Collapse
|
14
|
Gao Y, Basile JI, Classon C, Gavier-Widen D, Yoshimura A, Carow B, Rottenberg ME. STAT3 expression by myeloid cells is detrimental for the T- cell-mediated control of infection with Mycobacterium tuberculosis. PLoS Pathog 2018; 14:e1006809. [PMID: 29338039 PMCID: PMC5800682 DOI: 10.1371/journal.ppat.1006809] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 02/06/2018] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
STAT3 is a master regulator of the immune responses. Here we show that M. tuberculosis-infected stat3fl/fllysm cre mice, defective for STAT3 in myeloid cells, contained lower bacterial load in lungs and spleens, reduced granuloma extension but higher levels of pulmonary neutrophils. STAT3-deficient macrophages showed no improved control of intracellular mycobacterial growth. Instead, protection associated to elevated ability of stat3fl/fllysm cre antigen-presenting cells (APCs) to release IL-6 and IL-23 and to stimulate IL-17 secretion by mycobacteria-specific T cells. The increased IL-17 secretion accounted for the improved control of infection since neutralization of IL-17 receptor A in stat3fl/fllysm cre mice hampered bacterial control. APCs lacking SOCS3, which inhibits STAT3 activation via several cytokine receptors, were poor inducers of priming and of the IL-17 production by mycobacteria-specific T cells. In agreement, socs3fl/flcd11c cre mice deficient of SOCS3 in DCs showed increased susceptibility to M. tuberculosis infection. While STAT3 in APCs hampered IL-17 responses, STAT3 in mycobacteria-specific T cells was critical for IL-17 secretion, while SOCS3 in T cells impeded IL-17 secretion. Altogether, STAT3 signalling in myeloid cells is deleterious in the control of infection with M. tuberculosis. We studied the role of STAT3, a major regulator of immunity, in the control of the infection with M. tuberculosis. Stat3fl/fllysm cre mice, deficient in STAT3 in myeloid cells, showed lower bacterial levels in organs and reduced extension of lung granulomas after infection with M. tuberculosis. STAT3-deficient APCs stimulated with innate receptor agonists released high levels of IL-6 and IL-23, and promoted IL-17 production by mycobacteria-specific CD4+ T cells. Increased IL-17 levels accounted for the increased resistance to M. tuberculosis of the STAT3-deficient mice. Instead, stat3fl/fllysm cre macrophages showed no improved control of mycobacterial growth. SOCS3 is a negative regulator of STAT3 activation. The ability of socs3fl/fllysm cre APCs to secrete IL-6 and IL-23 and to stimulate IL-17 production by antigen-specific T cells was reduced. In agreement, mice lacking SOCS3 in DCs showed increased susceptibility to M. tuberculosis infection. Different to a role in myeloid cells, STAT3 expression by mycobacteria-specific T cells was required for IL-17 secretion while SOCS3 in T cells hampered IL-17 production. Therefore, despite STAT3 expression in T cells is required for Th17 differentiation, STAT3 in APCs hampers secretion of Th17 promoting cytokines and the secretion of IL-17 by mycobacteria-specific T cells and reduces the resistance of mice to infection with M. tuberculosis.
Collapse
Affiliation(s)
- Yu Gao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Ignacio Basile
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Cajsa Classon
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Dolores Gavier-Widen
- Department of Pathology and Wild Life Diseases, Swedish National Veterinary Institute, Uppsala, Sweden
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin E. Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
15
|
Duncan SA, Baganizi DR, Sahu R, Singh SR, Dennis VA. SOCS Proteins as Regulators of Inflammatory Responses Induced by Bacterial Infections: A Review. Front Microbiol 2017; 8:2431. [PMID: 29312162 PMCID: PMC5733031 DOI: 10.3389/fmicb.2017.02431] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/23/2017] [Indexed: 12/31/2022] Open
Abstract
Severe bacterial infections can lead to both acute and chronic inflammatory conditions. Innate immunity is the first defense mechanism employed against invading bacterial pathogens through the recognition of conserved molecular patterns on bacteria by pattern recognition receptors (PRRs), especially the toll-like receptors (TLRs). TLRs recognize distinct pathogen-associated molecular patterns (PAMPs) that play a critical role in innate immune responses by inducing the expression of several inflammatory genes. Thus, activation of immune cells is regulated by cytokines that use the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway and microbial recognition by TLRs. This system is tightly controlled by various endogenous molecules to allow for an appropriately regulated and safe host immune response to infections. Suppressor of cytokine signaling (SOCS) family of proteins is one of the central regulators of microbial pathogen-induced signaling of cytokines, principally through the inhibition of the activation of JAK/STAT signaling cascades. This review provides recent knowledge regarding the role of SOCS proteins during bacterial infections, with an emphasis on the mechanisms involved in their induction and regulation of antibacterial immune responses. Furthermore, the implication of SOCS proteins in diverse processes of bacteria to escape host defenses and in the outcome of bacterial infections are discussed, as well as the possibilities offered by these proteins for future targeted antimicrobial therapies.
Collapse
Affiliation(s)
- Skyla A Duncan
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Dieudonné R Baganizi
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Shree R Singh
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
16
|
Rauch KS, Hils M, Lupar E, Minguet S, Sigvardsson M, Rottenberg ME, Izcue A, Schachtrup C, Schachtrup K. Id3 Maintains Foxp3 Expression in Regulatory T Cells by Controlling a Transcriptional Network of E47, Spi-B, and SOCS3. Cell Rep 2017; 17:2827-2836. [PMID: 27974197 DOI: 10.1016/j.celrep.2016.11.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 10/17/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022] Open
Abstract
The transcription factor Foxp3 dominantly controls regulatory T (Treg) cell function, and only its continuous expression guarantees the maintenance of full Treg cell-suppressive capacity. However, transcriptional regulators maintaining Foxp3 transcription are incompletely described. Here, we report that high E47 transcription factor activity in Treg cells resulted in unstable Foxp3 expression. Under homeostatic conditions, Treg cells expressed high levels of the E47 antagonist Id3, thus restricting E47 activity and maintaining Foxp3 expression. In contrast, stimulation of Id3-deficient or E47-overexpressing Treg cells resulted in the loss of Foxp3 expression in a subset of Treg cells in vivo and in vitro. Mechanistic analysis indicated that E47 activated expression of the transcription factor Spi-B and the suppressor of cytokine signaling 3 (SOCS3), which both downregulated Foxp3 expression. These findings demonstrate that the balance of Id3 and E47 controls the maintenance of Foxp3 expression in Treg cells and, thus, contributes to Treg cell plasticity.
Collapse
Affiliation(s)
- Katharina S Rauch
- Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Miriam Hils
- Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Ekaterina Lupar
- Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Mikael Sigvardsson
- Institution for Clinical and Experimental Sciences, Faculty of Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ana Izcue
- Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Kristina Schachtrup
- Center for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
17
|
Schmok E, Abad Dar M, Behrends J, Erdmann H, Rückerl D, Endermann T, Heitmann L, Hessmann M, Yoshimura A, Rose-John S, Scheller J, Schaible UE, Ehlers S, Lang R, Hölscher C. Suppressor of Cytokine Signaling 3 in Macrophages Prevents Exacerbated Interleukin-6-Dependent Arginase-1 Activity and Early Permissiveness to Experimental Tuberculosis. Front Immunol 2017; 8:1537. [PMID: 29176982 PMCID: PMC5686055 DOI: 10.3389/fimmu.2017.01537] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/27/2017] [Indexed: 01/16/2023] Open
Abstract
Suppressor of cytokine signaling 3 (SOCS3) is a feedback inhibitor of interleukin (IL)-6 signaling in macrophages. In the absence of this molecule, macrophages become extremely prone to an IL-6-dependent expression of arginase-1 (Arg1) and nitric oxide synthase (NOS)2, the prototype markers for alternative or classical macrophage activation, respectively. Because both enzymes are antipodean macrophage effector molecules in Mycobacterium tuberculosis (Mtb) infection, we assessed the relevance of SOCS3 for macrophage activation during experimental tuberculosis using macrophage-specific SOCS3-deficient (LysMcreSOCS3loxP/loxP) mice. Aerosol infection of LysMcreSOCS3loxP/loxP mice resulted in remarkably higher bacterial loads in infected lungs and exacerbated pulmonary inflammation. This increased susceptibility to Mtb infection was accompanied by enhanced levels of both classical and alternative macrophage activation. However, high Arg1 expression preceded the increased induction of NOS2 and at early time points of infection mycobacteria were mostly found in cells positive for Arg1. This sequential activation of Arg1 and NOS2 expression in LysMcreSOCS3loxP/loxP mice appears to favor the initial replication of Mtb particularly in Arg1-positive cells. Neutralization of IL-6 in Mtb-infected LysMcreSOCS3loxP/loxP mice reduced arginase activity and restored control of mycobacterial replication in LysMcreSOCS3loxP/loxP mice. Our data reveal an unexpected role of SOCS3 during experimental TB: macrophage SOCS3 restrains early expression of Arg1 and helps limit Mtb replication in resident lung macrophages, thereby limiting the growth of mycobacteria. Together, SOCS3 keeps IL-6-dependent divergent macrophage responses such as Nos2 and Arg1 expression under control and safeguard protective macrophage effector mechanisms.
Collapse
Affiliation(s)
- Erik Schmok
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Mahin Abad Dar
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Jochen Behrends
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Hanna Erdmann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Dominik Rückerl
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Tanja Endermann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | - Lisa Heitmann
- Infection Immunology, Research Center Borstel, Borstel, Germany
| | | | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Graduate School of Medicine, Keio University, Tokyo, Japan
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-University, Kiel, Germany.,Cluster of Excellence Inflammation-at-Interfaces (Borstel-Kiel-Lübeck-Plön), Kiel, Germany
| | - Jürgen Scheller
- Medical Faculty, Institute of Biochemistry and Molecular Biology II, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Stefan Ehlers
- Cluster of Excellence Inflammation-at-Interfaces (Borstel-Kiel-Lübeck-Plön), Kiel, Germany.,Microbial Inflammation Research, Research Center Borstel, Borstel, Germany.,Molecular Inflammation Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Center Borstel, Borstel, Germany.,Cluster of Excellence Inflammation-at-Interfaces (Borstel-Kiel-Lübeck-Plön), Kiel, Germany
| |
Collapse
|
18
|
Fogelson SB, Fast MD, Leary J, Camus AC. Pathologic features of mycobacteriosis in naturally infected Syngnathidae and novel transcriptome assembly in association with disease. JOURNAL OF FISH DISEASES 2017; 40:1681-1694. [PMID: 28449243 DOI: 10.1111/jfd.12634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/11/2017] [Accepted: 02/12/2017] [Indexed: 06/07/2023]
Abstract
Syngnathidae (seahorses, seadragons and pipefish) suffer significant losses from non-tuberculous mycobacteria. However, they produce markedly different lesions in response to the disease compared to other teleost species, notably infrequent granuloma formation. This study evaluated 270 syngnathid fish, from which 92 were diagnosed with mycobacteriosis by histopathology, culture or both. Microscopic lesions variably consisted of random foci of coagulative necrosis in multiple organs, containing high numbers of free bacteria and large aggregates or sheets of macrophages with cytoplasm laden with acid-fast bacilli. Mycobacterial associated granulomas were identified in only six seahorses. Five fish had positive cultures with no observed microscopic changes. RNA-seq of the head kidney was performed to investigate the transcriptome of two infected and six non-infected lined seahorses Hippocampus erectus. Assembled and annotated putative transcripts serve to enrich the database for this species, as well as provide baseline data for understanding the pathogenesis of mycobacteriosis in seahorses. Putative components of the innate immune system (IL-1β, IL-6, TNF, NOS, Toll-like receptor 1, MHC Class I, NF-κβ, transforming growth factor beta, MyD88) were identified in the RNA-seq data set. However, a homolog for a key component in the TH1 adaptive immune response, interferon-gamma, was not identified and may underlie the unique pathologic presentation.
Collapse
Affiliation(s)
- S B Fogelson
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - M D Fast
- Department of Pathology and Microbiology, University of Prince Edward Island, Atlantic Veterinary College, Charlottetown, PE, Canada
| | - J Leary
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - A C Camus
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
19
|
Carow B, Gao Y, Terán G, Yang XO, Dong C, Yoshimura A, Rottenberg ME. CISH controls bacterial burden early after infection with Mycobacterium tuberculosis in mice. Tuberculosis (Edinb) 2017; 107:175-180. [PMID: 29050767 DOI: 10.1016/j.tube.2017.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/08/2017] [Accepted: 09/17/2017] [Indexed: 10/18/2022]
Abstract
CISH gene has been associated with increased susceptibility to human tuberculosis. We found that cish-/- mice had higher M. tuberculosis load in spleens and lungs up to 2.5 weeks after infection but not later compared to controls. Cish mRNA levels were increased in lungs at early and late time points after M. tuberculosis infection. In relation, the titers of inos and tnf mRNA in lungs were reduced early after infection of cish-/- mice. The transfer of cish-/- and control T cells conferred rag1-/- mice similar protection to infection with M. tuberculosis. Macrophages showed increased cish mRNA levels after M. tuberculosis infection in vitro. However, mycobacterial uptake and growth in cish-/- and control macrophages was similar. Thus, we here show that CISH mediates control of M. tuberculosis in mice early after infection via regulation of innate immune mechanisms.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yu Gao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Graciela Terán
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; Immunoparasitology Unit, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
20
|
Sogi KM, Lien KA, Johnson JR, Krogan NJ, Stanley SA. The Tyrosine Kinase Inhibitor Gefitinib Restricts Mycobacterium tuberculosis Growth through Increased Lysosomal Biogenesis and Modulation of Cytokine Signaling. ACS Infect Dis 2017; 3:564-574. [PMID: 28537707 PMCID: PMC5728371 DOI: 10.1021/acsinfecdis.7b00046] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Host-directed therapeutics have the potential to combat the global tuberculosis pandemic. We previously identified gefitinib, an inhibitor of EGFR, as a potential host-targeted therapeutic effective against Mycobacterium tuberculosis infection of macrophages and mice. Here we examine the functional consequences of gefitinib treatment on M. tuberculosis infected macrophages. Using phosphoproteomic and transcriptional profiling, we identify two mechanisms by which gefitinib influences macrophage responses to infection to affect cytokine responses and limit replication of M. tuberculosis in macrophages. First, we find that gefitinib treatment of M. tuberculosis infected macrophages inhibits STAT3, a transcription factor known to repress effective immune responses to M. tuberculosis in vivo. Second, we find that gefitinib treatment of M. tuberculosis infected macrophages leads to increased expression of genes involved in lysosomal biogenesis and function and an increase of functional lysosomes in gefitinib treated cells. Furthermore, we show that gefitinib treatment increases the targeting of bacteria to lysosomes, providing an explanation for the cell intrinsic effects of gefitinib treatment on M. tuberculosis infection. Our data provide novel insights into the effects of gefitinib on mammalian cells and into the possible roles for EGFR signaling in macrophages.
Collapse
Affiliation(s)
- Kimberly M. Sogi
- School of Public Health, Division of Infectious Diseases and Vaccinology, Li Ka Shing Center, University of California, Berkeley, California 94720, United States
| | - Katie A. Lien
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Jeffrey R. Johnson
- Department of Cellular and Molecular Pharmacology, University of California—San Francisco, San Francisco, California 94158, United States
| | - Nevan J. Krogan
- Department of Cellular and Molecular Pharmacology, University of California—San Francisco, San Francisco, California 94158, United States
| | - Sarah A. Stanley
- School of Public Health, Division of Infectious Diseases and Vaccinology, Li Ka Shing Center, University of California, Berkeley, California 94720, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| |
Collapse
|
21
|
Jung BG, Wang X, Yi N, Ma J, Turner J, Samten B. Early Secreted Antigenic Target of 6-kDa of Mycobacterium tuberculosis Stimulates IL-6 Production by Macrophages through Activation of STAT3. Sci Rep 2017; 7:40984. [PMID: 28106119 PMCID: PMC5247711 DOI: 10.1038/srep40984] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/13/2016] [Indexed: 01/08/2023] Open
Abstract
As early secreted antigenic target of 6 kDa (ESAT-6) of Mycobacterium tuberculosis (Mtb) is an essential virulence factor and macrophages are critical for tuberculosis infection and immunity, we studied ESAT-6 stimulated IL-6 production by macrophages. ESAT-6 stimulated significantly higher IL-6 secretion by murine bone marrow derived macrophages (BMDM) compared to culture filtrate protein 10 kDa (CFP10) and antigen 85A. Polymyxin B, an LPS blocker, did not affect ESAT-6 stimulated macrophage IL-6 production. ESAT-6 but not Pam3CSK4 induced IL-6 by TLR2 knockout BMDM. ESAT-6 induced phosphorylation and DNA binding of STAT3 and this was blocked by STAT3 inhibitors but not by rapamycin. STAT3 inhibitors suppressed ESAT-6-induced IL-6 transcription and secretion without affecting cell viability. This was confirmed by silencing STAT3 in macrophages. Blocking neither IL-6Rα/IL-6 nor IL-10 affected ESAT-6-induced STAT3 activation and IL-6 production. Infection of BMDM and human macrophages with Mtb with esat-6 deletion induced diminished STAT3 activation and reduced IL-6 production compared to wild type and esat-6 complemented Mtb strains. Administration of ESAT-6 but not CFP10 induced STAT3 phosphorylation and IL-6 expression in the mouse lungs, consistent with expression of ESAT-6, IL-6 and phosphorylated-STAT3 in Mtb-infected mouse lungs. We conclude that ESAT-6 stimulates macrophage IL-6 production through STAT3 activation.
Collapse
Affiliation(s)
- Bock-Gie Jung
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Texas 75708, USA
| | - Xisheng Wang
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Texas 75708, USA
| | - Na Yi
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Texas 75708, USA
| | - Justin Ma
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Texas 75708, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity and Center for Microbial Interface Biology, the Ohio State University, Columbus, OH 43210, USA
| | - Buka Samten
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Texas 75708, USA
| |
Collapse
|
22
|
Mahony R, Ahmed S, Diskin C, Stevenson NJ. SOCS3 revisited: a broad regulator of disease, now ready for therapeutic use? Cell Mol Life Sci 2016; 73:3323-36. [PMID: 27137184 PMCID: PMC11108554 DOI: 10.1007/s00018-016-2234-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/24/2016] [Accepted: 04/19/2016] [Indexed: 12/17/2022]
Abstract
Since their discovery, SOCS have been characterised as regulatory cornerstones of intracellular signalling. While classically controlling the JAK/STAT pathway, their inhibitory effects are documented across several cascades, underpinning their essential role in homeostatic maintenance and disease. After 20 years of extensive research, SOCS3 has emerged as arguably the most important family member, through its regulation of both cytokine- and pathogen-induced cascades. In fact, low expression of SOCS3 is associated with autoimmunity and oncogenesis, while high expression is linked to diabetes and pathogenic immune evasion. The induction of SOCS3 by both viruses and bacteria and its impact upon inflammatory disorders, underscores this protein's increasing clinical potential. Therefore, with the aim of highlighting SOCS3 as a therapeutic target for future development, this review revisits its multi-faceted immune regulatory functions and summarises its role in a broad ranges of diseases.
Collapse
Affiliation(s)
- R Mahony
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - S Ahmed
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - C Diskin
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - N J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
23
|
Carion TW, McWhirter CR, Grewal DK, Berger EA. Efficacy of VIP as Treatment for Bacteria-Induced Keratitis Against Multiple Pseudomonas aeruginosa Strains. Invest Ophthalmol Vis Sci 2016; 56:6932-40. [PMID: 26513498 DOI: 10.1167/iovs.15-17315] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Previous studies have demonstrated the efficacy of vasoactive intestinal peptide (VIP) treatment in regulating inflammation following bacterial keratitis induced by the P. aeruginosa strain 19660. However, in the current study we assessed whether disease outcome is specific to 19660 or if VIP treatment is effective against multiple P. aeruginosa strains. METHODS B6 mice received daily IP injections of VIP from -1 through 5 days post injection (p.i.). Control mice were similarly injected with PBS. Corneal infection was induced using PA 19660, PAO1 or KEI 1025. Disease response was documented and bacterial plate counts and myeloperoxidase assays were performed. Expression of select inflammatory mediators as well as enzymes associated with lipid mediator production was assessed after VIP treatment. KEI 1025 was characterized by cytotoxicity and invasion assays and then confirmed for ExoS/ExoU expression. RESULTS VIP treatment converted the susceptible response to resistant for the three P. aeruginosa strains tested. Disease response was significantly reduced with no corneal perforation. Anti-inflammatory mediators were enhanced after VIP treatment, while pro-inflammatory molecules were reduced compared to controls. Furthermore, VIP reduced inflammatory cell persistence in the cornea after infection with each of the P. aeruginosa strains. CONCLUSIONS VIP treatment is effective at ameliorating disease pathogenesis for multiple P. aeruginosa strains, both cytotoxic and invasive. This study is also the first to indicate a possible role for VIP regarding lipid mediator expression in the eye. In addition, the clinical isolate, KEI 1025, was characterized as an invasive strain. Overall, this study strengthens the preclinical development of VIP as a therapeutic agent for ocular infectious disease.
Collapse
Affiliation(s)
- Thomas W Carion
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Cody R McWhirter
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Daiyajot K Grewal
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Elizabeth A Berger
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States 2Department of Ophthalmology, Kresge Eye Institute, Detroit, Michigan, United States
| |
Collapse
|
24
|
Tuberculosis is associated with expansion of a motile, permissive and immunomodulatory CD16(+) monocyte population via the IL-10/STAT3 axis. Cell Res 2015; 25:1333-51. [PMID: 26482950 DOI: 10.1038/cr.2015.123] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/31/2015] [Accepted: 09/06/2015] [Indexed: 01/23/2023] Open
Abstract
The human CD14(+) monocyte compartment is composed by two subsets based on CD16 expression. We previously reported that this compartment is perturbed in tuberculosis (TB) patients, as reflected by the expansion of CD16(+) monocytes along with disease severity. Whether this unbalance is beneficial or detrimental to host defense remains to be elucidated. Here in the context of active TB, we demonstrate that human monocytes are predisposed to differentiate towards an anti-inflammatory (M2-like) macrophage activation program characterized by the CD16(+)CD163(+)MerTK(+)pSTAT3(+) phenotype and functional properties such as enhanced protease-dependent motility, pathogen permissivity and immunomodulation. This process is dependent on STAT3 activation, and loss-of-function experiments point towards a detrimental role in host defense against TB. Importantly, we provide a critical correlation between the abundance of the CD16(+)CD163(+)MerTK(+)pSTAT3(+) cells and the progression of the disease either at the local level in a non-human primate tuberculous granuloma context, or at the systemic level through the detection of the soluble form of CD163 in human sera. Collectively, this study argues for the pathogenic role of the CD16(+)CD163(+)MerTK(+)pSTAT3(+) monocyte-to-macrophage differentiation program and its potential as a target for TB therapy, and promotes the detection of circulating CD163 as a potential biomarker for disease progression and monitoring of treatment efficacy.
Collapse
|
25
|
Saybani MR, Shamshirband S, Golzari Hormozi S, Wah TY, Aghabozorgi S, Pourhoseingholi MA, Olariu T. Diagnosing tuberculosis with a novel support vector machine-based artificial immune recognition system. IRANIAN RED CRESCENT MEDICAL JOURNAL 2015; 17:e24557. [PMID: 26023340 PMCID: PMC4443397 DOI: 10.5812/ircmj.17(4)2015.24557] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/28/2015] [Accepted: 02/07/2015] [Indexed: 12/04/2022]
Abstract
BACKGROUND Tuberculosis (TB) is a major global health problem, which has been ranked as the second leading cause of death from an infectious disease worldwide. Diagnosis based on cultured specimens is the reference standard, however results take weeks to process. Scientists are looking for early detection strategies, which remain the cornerstone of tuberculosis control. Consequently there is a need to develop an expert system that helps medical professionals to accurately and quickly diagnose the disease. Artificial Immune Recognition System (AIRS) has been used successfully for diagnosing various diseases. However, little effort has been undertaken to improve its classification accuracy. OBJECTIVES In order to increase the classification accuracy of AIRS, this study introduces a new hybrid system that incorporates a support vector machine into AIRS for diagnosing tuberculosis. PATIENTS AND METHODS Patient epacris reports obtained from the Pasteur laboratory of Iran were used as the benchmark data set, with the sample size of 175 (114 positive samples for TB and 60 samples in the negative group). The strategy of this study was to ensure representativeness, thus it was important to have an adequate number of instances for both TB and non-TB cases. The classification performance was measured through 10-fold cross-validation, Root Mean Squared Error (RMSE), sensitivity and specificity, Youden's Index, and Area Under the Curve (AUC). Statistical analysis was done using the Waikato Environment for Knowledge Analysis (WEKA), a machine learning program for windows. RESULTS With an accuracy of 100%, sensitivity of 100%, specificity of 100%, Youden's Index of 1, Area Under the Curve of 1, and RMSE of 0, the proposed method was able to successfully classify tuberculosis patients. CONCLUSIONS There have been many researches that aimed at diagnosing tuberculosis faster and more accurately. Our results described a model for diagnosing tuberculosis with 100% sensitivity and 100% specificity. This model can be used as an additional tool for experts in medicine to diagnose TBC more accurately and quickly.
Collapse
Affiliation(s)
- Mahmoud Reza Saybani
- Department of Information Systems, Faculty of Computer Science and Information Technology, University of Malaya, Kula Lumpur, Malaysia
| | | | - Shahram Golzari Hormozi
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Hormozgan, Bandar Abbas, IR Iran
| | - Teh Ying Wah
- Department of Information Systems, Faculty of Computer Science and Information Technology, University of Malaya, Kula Lumpur, Malaysia
| | - Saeed Aghabozorgi
- Department of Information Systems, Faculty of Computer Science and Information Technology, University of Malaya, Kula Lumpur, Malaysia
| | - Mohamad Amin Pourhoseingholi
- Gastroenterology and Liver Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Teodora Olariu
- Department of Intensive Care, Faculty of Medicine, Western University of Arad, Arad, Romania
| |
Collapse
|
26
|
Blok DC, Kager LM, Hoogendijk AJ, Lede IO, Rahman W, Afroz R, Bresser P, van der Zee JS, Ghose A, Visser CE, de Jong MD, Zahed ASM, Husain MA, Alam KM, Barua PC, Hassan M, Hossain A, Tayab MA, Lutter R, Day N, Dondorp AM, de Vos AF, van 't Veer C, van der Poll T. Expression of inhibitory regulators of innate immunity in patients with active tuberculosis. BMC Infect Dis 2015; 15:98. [PMID: 25887604 PMCID: PMC4365962 DOI: 10.1186/s12879-015-0833-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 02/12/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Tuberculosis (TB) is an important cause of morbidity and mortality worldwide. Toll-like-receptors (TLRs) are important for the recognition of the causative agent Mycobacterium tuberculosis. Negative regulation of TLRs is necessary to control deleterious inflammatory damage, but could provide a means of immune evasion by M. tuberculosis as well. METHODS To obtain insight in the extent of expression of inhibitory regulators of immunity in patients with active TB, peripheral-blood-mononuclear-cells (PBMCs) and plasma were obtained from 54 TB patients and 29 healthy blood donors from Chittagong, Bangladesh. Bilateral alveolar macrophages were obtained from an infected versus a contralateral normal lung segment of 9 patients. Statistical analyses were performed using Mann-Whitney U and Wilcoxon matched pairs testing. Correlations were calculated using the Spearman rho test. RESULTS PBMCs harvested from TB patients demonstrated increased mRNA expression of IL-1-receptor-associated-kinase-M, suppressor-of-cytokine-signalling-3 and Toll-interacting-protein. Flow cytometry revealed enhanced expression of IL-1-receptor-like-1 (ST2) on lymphocytes. Plasma soluble ST2 was elevated in patients with TB and correlated with established TB biomarkers, most strongly with soluble interleukin-2 receptor subunit α and interleukin-8. Alveolar macrophage mRNA expression of negative TLR regulators did not differ between the infected and contralateral lung side. CONCLUSION These results show enhanced expression of distinct negative regulators of innate immunity in PBMCs of patients with TB and identify plasma soluble ST2 as a potential novel biomarker for TB disease activity.
Collapse
Affiliation(s)
- Dana C Blok
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Liesbeth M Kager
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Arie J Hoogendijk
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Ivar O Lede
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Department of Microbiology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Wahid Rahman
- Department of Internal Medicine, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Rumana Afroz
- Department of Internal Medicine, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Paul Bresser
- Department of Pulmonology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Department of Pulmonology, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Jaring S van der Zee
- Department of Pulmonology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Department of Pulmonology, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Aniruddha Ghose
- Department of Internal Medicine, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Caroline E Visser
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Department of Microbiology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Menno D de Jong
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Department of Microbiology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Abu Shahed Md Zahed
- Department of Internal Medicine, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Md Anwar Husain
- Department of Microbiology, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Khan Mashrequl Alam
- Department of Microbiology, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | | | - Mahtabuddin Hassan
- Department of Internal Medicine, Chittagong Medical College and Hospital (CMCH), Chittagong, Bangladesh.
| | - Ahmed Hossain
- Chest Disease Clinic Chittagong (CDCC), Chittagong, Bangladesh
| | - Md Abu Tayab
- Chittagong General Hospital, Chittagong, Bangladesh.
| | - Rene Lutter
- Department of Pulmonology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Department of Experimental Immunology, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands.
| | - Nick Day
- Mahidol Oxford Tropical Research Unit, Mahidol University, Bangkok, Thailand.
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Research Unit, Mahidol University, Bangkok, Thailand.
| | - Alex F de Vos
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Cornelis van 't Veer
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| | - Tom van der Poll
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands. .,Division of Infectious Diseases, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Rottenberg ME, Carow B. SOCS3 and STAT3, major controllers of the outcome of infection with Mycobacterium tuberculosis. Semin Immunol 2014; 26:518-32. [DOI: 10.1016/j.smim.2014.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 01/04/2023]
|
28
|
Rajaram MVS, Ni B, Dodd CE, Schlesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Semin Immunol 2014; 26:471-85. [PMID: 25453226 DOI: 10.1016/j.smim.2014.09.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/17/2022]
Abstract
Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs).
Collapse
Affiliation(s)
- Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Ni
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Claire E Dodd
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
29
|
Ozkan ZS, Deveci D, Kumbak B, Simsek M, Ilhan F, Sekercioglu S, Sapmaz E. What is the impact of Th1/Th2 ratio, SOCS3, IL17, and IL35 levels in unexplained infertility? J Reprod Immunol 2014; 103:53-8. [DOI: 10.1016/j.jri.2013.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/10/2013] [Accepted: 11/14/2013] [Indexed: 12/15/2022]
|
30
|
Ozkan ZS, Deveci D, Simsek M, Ilhan F, Risvanli A, Sapmaz E. What is the impact of SOCS3, IL-35 and IL17 in immune pathogenesis of recurrent pregnancy loss? J Matern Fetal Neonatal Med 2014; 28:324-8. [PMID: 24762139 DOI: 10.3109/14767058.2014.916676] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To investigate the plasma levels of interleukin-4 (IL-4), IL-6, IL-10, tumor necrosis factor-alpha (TNF-alpha), interferon-gamma (IFN-gamma), transforming growth factor-beta (TGF-beta), IL-17, IL-35 and suppressor of cytokine signaling 3 (SOCS3) in the women with history of idiopathic recurrent pregnancy loss (RPL) and in the fertile controls. METHODS This study was conducted with 60 idiopathic RPL cases and 40 age-matched fertile controls. Mid-follicular plasma levels of IL-17, IFN-gamma, TNF-alpha, TGF-beta, IL-6, IL-4, IL-10, SOCS3 and IL-35 were assayed by an enzyme linked immunosorbent assay. RESULTS The mean age of RPL and control cases were 31.6 ± 0.6 and 32.1 ± 0.7 years, respectively. While plasma IL-35 and SOCS3 levels of RPL group were significantly lower than that of the control group; IFN-gamma, TNF-alpha, IL-4, IL-6, IL-10, IL-17 and TGF-beta levels of RPL group were significantly higher than that of the control group. The comparison of cytokine ratios between RPL and control groups indicated significantly high TNF-alpha/IL-10, TNF-alpha/IL-4, IFN-gamma/IL-10, IFN-gamma/IL-6 and IFN-gamma/IL-4 ratios in the RPL group. IL-35/IL-17 ratio was significantly low in the RPL group compared to that in the control group. Overstimulation of TNF-alpha presented moderate influence on recurrent miscarriage risk. CONCLUSION Decreased SOCS3 and IL-35 plasma levels and increased Th1/Th2 cytokine ratios in RPL cases pointed out the supression of anti-inflammatory process and this supression might play an important role in the pathogenesis of idiopathic RPL.
Collapse
Affiliation(s)
- Zehra Sema Ozkan
- Department of Obstetrics and Gynecology, Firat University School of Medicine , Elazig , Turkey
| | | | | | | | | | | |
Collapse
|
31
|
Progress in tuberculosis vaccine development and host-directed therapies--a state of the art review. THE LANCET RESPIRATORY MEDICINE 2014; 2:301-20. [PMID: 24717627 DOI: 10.1016/s2213-2600(14)70033-5] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tuberculosis continues to kill 1·4 million people annually. During the past 5 years, an alarming increase in the number of patients with multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis has been noted, particularly in eastern Europe, Asia, and southern Africa. Treatment outcomes with available treatment regimens for drug-resistant tuberculosis are poor. Although substantial progress in drug development for tuberculosis has been made, scientific progress towards development of interventions for prevention and improvement of drug treatment outcomes have lagged behind. Innovative interventions are therefore needed to combat the growing pandemic of multidrug-resistant and extensively drug-resistant tuberculosis. Novel adjunct treatments are needed to accomplish improved cure rates for multidrug-resistant and extensively drug-resistant tuberculosis. A novel, safe, widely applicable, and more effective vaccine against tuberculosis is also desperately sought to achieve disease control. The quest to develop a universally protective vaccine for tuberculosis continues. So far, research and development of tuberculosis vaccines has resulted in almost 20 candidates at different stages of the clinical trial pipeline. Host-directed therapies are now being developed to refocus the anti-Mycobacterium tuberculosis-directed immune responses towards the host; a strategy that could be especially beneficial for patients with multidrug-resistant tuberculosis or extensively drug-resistant tuberculosis. As we are running short of canonical tuberculosis drugs, more attention should be given to host-directed preventive and therapeutic intervention measures.
Collapse
|
32
|
Carow B, Rottenberg ME. SOCS3, a Major Regulator of Infection and Inflammation. Front Immunol 2014; 5:58. [PMID: 24600449 PMCID: PMC3928676 DOI: 10.3389/fimmu.2014.00058] [Citation(s) in RCA: 399] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/31/2014] [Indexed: 12/18/2022] Open
Abstract
In this review, we describe the role of suppressor of cytokine signaling-3 (SOCS3) in modulating the outcome of infections and autoimmune diseases as well as the underlying mechanisms. SOCS3 regulates cytokine or hormone signaling usually preventing, but in some cases aggravating, a variety of diseases. A main role of SOCS3 results from its binding to both the JAK kinase and the cytokine receptor, which results in the inhibition of STAT3 activation. Available data also indicate that SOCS3 can regulate signaling via other STATs than STAT3 and also controls cellular pathways unrelated to STAT activation. SOCS3 might either act directly by hampering JAK activation or by mediating the ubiquitination and subsequent proteasome degradation of the cytokine/growth factor/hormone receptor. Inflammation and infection stimulate SOCS3 expression in different myeloid and lymphoid cell populations as well as in diverse non-hematopoietic cells. The accumulated data suggest a relevant program coordinated by SOCS3 in different cell populations, devoted to the control of immune homeostasis in physiological and pathological conditions such as infection and autoimmunity.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
33
|
Ashenafi S, Aderaye G, Bekele A, Zewdie M, Aseffa G, Hoang ATN, Carow B, Habtamu M, Wijkander M, Rottenberg M, Aseffa A, Andersson J, Svensson M, Brighenti S. Progression of clinical tuberculosis is associated with a Th2 immune response signature in combination with elevated levels of SOCS3. Clin Immunol 2014; 151:84-99. [PMID: 24584041 DOI: 10.1016/j.clim.2014.01.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/08/2014] [Accepted: 01/30/2014] [Indexed: 02/06/2023]
Abstract
In this study, we explored the local cytokine/chemokine profiles in patients with active pulmonary or pleural tuberculosis (TB) using multiplex protein analysis of bronchoalveolar lavage and pleural fluid samples. Despite increased pro-inflammation compared to the uninfected controls; there was no up-regulation of IFN-γ or the T cell chemoattractant CCL5 in the lung of patients with pulmonary TB. Instead, elevated levels of IL-4 and CCL4 were associated with high mycobacteria-specific IgG titres as well as SOCS3 (suppressors of cytokine signaling) mRNA and progression of moderate-to-severe disease. Contrary, IL-4, CCL4 and SOCS3 remained low in patients with extrapulmonary pleural TB, while IFN-γ, CCL5 and SOCS1 were up-regulated. Both SOCS molecules were induced in human macrophages infected with Mycobacterium tuberculosis in vitro. The Th2 immune response signature found in patients with progressive pulmonary TB could result from inappropriate cytokine/chemokine responses and excessive SOCS3 expression that may represent potential targets for clinical TB management.
Collapse
Affiliation(s)
- Senait Ashenafi
- Karolinska Institutet, Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden; Black Lion University Hospital and Addis Ababa University, Department of Pathology, Faculty of Medicine, Addis Ababa, Ethiopia
| | - Getachew Aderaye
- Black Lion University Hospital and Addis Ababa University, Department of Internal Medicine, Faculty of Medicine, Addis Ababa, Ethiopia
| | - Amsalu Bekele
- Black Lion University Hospital and Addis Ababa University, Department of Internal Medicine, Faculty of Medicine, Addis Ababa, Ethiopia
| | - Martha Zewdie
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Getachew Aseffa
- Black Lion University Hospital and Addis Ababa University, Department of Radiology, Faculty of Medicine, Addis Ababa, Ethiopia
| | - Anh Thu Nguyen Hoang
- Karolinska Institutet, Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Berit Carow
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology (MTC), Stockholm, Sweden
| | - Meseret Habtamu
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Maria Wijkander
- Karolinska Institutet, Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Martin Rottenberg
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology (MTC), Stockholm, Sweden
| | - Abraham Aseffa
- Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Jan Andersson
- Karolinska Institutet, Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden; Karolinska University Hospital Huddinge, Department of Medicine, Division of Infectious Diseases, Stockholm, Sweden
| | - Mattias Svensson
- Karolinska Institutet, Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Susanna Brighenti
- Karolinska Institutet, Center for Infectious Medicine (CIM), F59, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
34
|
Ozkan ZS, Simsek M, Ilhan F, Deveci D, Godekmerdan A, Sapmaz E. Plasma IL-17, IL-35, interferon-γ, SOCS3 and TGF-β levels in pregnant women with preeclampsia, and their relation with severity of disease. J Matern Fetal Neonatal Med 2013; 27:1513-7. [PMID: 24175856 DOI: 10.3109/14767058.2013.861415] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
UNLABELLED Abstract Objective: To research the hypothesis of preeclampsia (PE) is associated with increased systemic inflammatory responses of Th1-type as well as decreased Th2-type responses; we evaluated the maternal plasma levels of IFN-gamma, TNF-alpha, TGF-beta, IL-4, IL-6, IL-10, IL-17, IL-35 and SOCS3 in preeclamptic and healthy pregnants. METHODS This study was conducted with 40 preeclamptic (study group) and 40 normotensive pregnant (control) women in third trimester when they were admitted to the labor and delivery unit. The extracted maternal plasma samples were assayed by an enzyme-linked immunosorbent assay. Statistical analysis was performed by SPSS 16.0 version. RESULTS While IFN-gamma and TGF-beta levels of preeclamptic women were significantly higher (p < 0.01), IL-35 and IL-17 levels of preeclamptic women were significantly lower (p < 0.01) than those of controls. The ratios of IFN-gamma/IL-10, IFN-gamma/IL-6, IFN-gamma/IL-4 were significantly high and ratio of IL-35/IL-17 was significantly low in the PE group compared to those in the control group. Maternal plasma SOCS3 levels showed negative correlation with blood pressure and proteinuria severity, but none of the cytokines showed influence on blood pressure and proteinuria after adjusting for maternal and gestational age. CONCLUSIONS Increased IFN-gamma/TGF-beta production and reduced IL-35/IL-17/SOCS3 production in preeclamptic women may lead to less cytokine inhibitory activity in PE, which may account for the increased proteinuria and blood pressure in PE.
Collapse
|