1
|
Bai X, Gao J, Guan X, Narum DE, Fornis LB, Griffith DE, Gao B, Sandhaus RA, Huang H, Chan ED. Analysis of alpha-1-antitrypsin (AAT)-regulated, glucocorticoid receptor-dependent genes in macrophages reveals a novel host defense function of AAT. Physiol Rep 2024; 12:e16124. [PMID: 39016119 PMCID: PMC11252833 DOI: 10.14814/phy2.16124] [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: 04/01/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/18/2024] Open
Abstract
Alpha-1-antitrypsin (AAT) plays a homeostatic role in attenuating excessive inflammation and augmenting host defense against microbes. We demonstrated previously that AAT binds to the glucocorticoid receptor (GR) resulting in significant anti-inflammatory and antimycobacterial consequences in macrophages. Our current investigation aims to uncover AAT-regulated genes that rely on GR in macrophages. We incubated control THP-1 cells (THP-1control) and THP-1 cells knocked down for GR (THP-1GR-KD) with AAT, performed bulk RNA sequencing, and analyzed the findings. In THP-1control cells, AAT significantly upregulated 408 genes and downregulated 376 genes. Comparing THP-1control and THP-1GR-KD cells, 125 (30.6%) of the AAT-upregulated genes and 154 (41.0%) of the AAT-downregulated genes were significantly dependent on GR. Among the AAT-upregulated, GR-dependent genes, CSF-2 that encodes for granulocyte-monocyte colony-stimulating factor (GM-CSF), known to be host-protective against nontuberculous mycobacteria, was strongly upregulated by AAT and dependent on GR. We further quantified the mRNA and protein of several AAT-upregulated, GR-dependent genes in macrophages and the mRNA of several AAT-downregulated, GR-dependent genes. We also discussed the function(s) of selected AAT-regulated, GR-dependent gene products largely in the context of mycobacterial infections. In conclusion, AAT regulated several genes that are dependent on GR and play roles in host immunity against mycobacteria.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of MedicineRocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Junfeng Gao
- Department of Immunology and Genomic MedicineNational Jewish HealthDenverColoradoUSA
| | - Xiaoyu Guan
- Department of Biostatistics and InformaticsUniversity of Colorado School of Public Health Anschutz Medical CampusAuroraColoradoUSA
| | - Drew E. Narum
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
| | | | - David E. Griffith
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineNational Jewish HealthDenverColoradoUSA
| | - Bifeng Gao
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Robert A. Sandhaus
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineNational Jewish HealthDenverColoradoUSA
| | - Hua Huang
- Department of Immunology and Genomic MedicineNational Jewish HealthDenverColoradoUSA
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Edward D. Chan
- Department of MedicineRocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
- Department of Academic AffairsNational Jewish HealthDenverColoradoUSA
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
2
|
Calcagno A, Coppola N, Sarmati L, Tadolini M, Parrella R, Matteelli A, Riccardi N, Trezzi M, Di Biagio A, Pirriatore V, Russo A, Gualano G, Pontali E, Surace L, Falbo E, Mencarini J, Palmieri F, Gori A, Schiuma M, Lapadula G, Goletti D. Drugs for treating infections caused by non-tubercular mycobacteria: a narrative review from the study group on mycobacteria of the Italian Society of Infectious Diseases and Tropical Medicine. Infection 2024; 52:737-765. [PMID: 38329686 PMCID: PMC11142973 DOI: 10.1007/s15010-024-02183-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Non-tuberculous mycobacteria (NTM) are generally free-living organism, widely distributed in the environment, with sporadic potential to infect. In recent years, there has been a significant increase in the global incidence of NTM-related disease, spanning across all continents and an increased mortality after the diagnosis has been reported. The decisions on whether to treat or not and which drugs to use are complex and require a multidisciplinary approach as well as patients' involvement in the decision process. METHODS AND RESULTS This review aims at describing the drugs used for treating NTM-associated diseases emphasizing the efficacy, tolerability, optimization strategies as well as possible drugs that might be used in case of intolerance or resistance. We also reviewed data on newer compounds highlighting the lack of randomised clinical trials for many drugs but also encouraging preliminary data for others. We also focused on non-pharmacological interventions that need to be adopted during care of individuals with NTM-associated diseases CONCLUSIONS: Despite insufficient efficacy and poor tolerability this review emphasizes the improvement in patients' care and the needs for future studies in the field of anti-NTM treatments.
Collapse
Affiliation(s)
- A Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy.
- Stop TB Italy, Milan, Italy.
| | - N Coppola
- Infectious Diseases Unit, Section of Infectious Diseases, Department of Mental Health and Public Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - L Sarmati
- Department of System Medicine, Tor Vergata University and Infectious Disease Clinic, Policlinico Tor Vergata, Rome, Italy
| | - M Tadolini
- Stop TB Italy, Milan, Italy
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - R Parrella
- Stop TB Italy, Milan, Italy
- Respiratory Infectious Diseases Unit, Cotugno Hospital, A. O. R. N. dei Colli, Naples, Italy
| | - A Matteelli
- Institute of Infectious and Tropical Diseases, WHO Collaborating Centre for TB Prevention, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - N Riccardi
- Stop TB Italy, Milan, Italy
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | - M Trezzi
- Stop TB Italy, Milan, Italy
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - A Di Biagio
- Infectious Diseases Unit, San Martino Policlinico Hospital-IRCCS for Oncology and Neurosciences, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - V Pirriatore
- Stop TB Italy, Milan, Italy
- Unit of Infectious Diseases, "DivisioneA", Ospedale Amedeo di Savoia, ASL CIttà di Torino, Turin, Italy
| | - A Russo
- Infectious Diseases Unit, Section of Infectious Diseases, Department of Mental Health and Public Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - G Gualano
- Stop TB Italy, Milan, Italy
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - E Pontali
- Department of Infectious Diseases, Galliera Hospital, Genoa, Italy
| | - L Surace
- Stop TB Italy, Milan, Italy
- Dipartimento Di Prevenzione, Azienda Sanitaria Provinciale di Catanzaro, Centro di Medicina del Viaggiatore e delle Migrazioni, P. O. Giovanni Paolo II, Lamezia Terme, CZ, Italy
| | - E Falbo
- Stop TB Italy, Milan, Italy
- Dipartimento Di Prevenzione, Azienda Sanitaria Provinciale di Catanzaro, Centro di Medicina del Viaggiatore e delle Migrazioni, P. O. Giovanni Paolo II, Lamezia Terme, CZ, Italy
| | - J Mencarini
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - F Palmieri
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - A Gori
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, ASST Fatebenefratelli Sacco-Ospedale Luigi Sacco-Polo Universitario and Università Degli Studi di Milano, Milano, Italy
| | - M Schiuma
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, ASST Fatebenefratelli Sacco-Ospedale Luigi Sacco-Polo Universitario and Università Degli Studi di Milano, Milano, Italy
| | - G Lapadula
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca, Monza, Italy
| | - D Goletti
- Stop TB Italy, Milan, Italy
- Translational Research Unit, Epidemiology Department, National Institute for Infectious Diseases-IRCCS L. Spallanzani, Rome, Italy
| |
Collapse
|
3
|
Ahmad S, Ahmed J, Khalifa EH, Khattak FA, Khan AS, Farooq SU, Osman SMA, Salih MM, Ullah N, Khan TA. Novel mutations in genes of the IL-12/IFN-γ axis cause susceptibility to tuberculosis. J Infect Public Health 2023; 16:1368-1378. [PMID: 37437430 DOI: 10.1016/j.jiph.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/15/2023] [Accepted: 06/06/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The IL-12/23/ISG15-IFN-γ pathway is the main immunological pathway for controlling intra-macrophagic microorganisms such as Mycobacteria, Salmonella, and Leishmania spp. Consequently, upon mutations in genes of the IL-12/23/ISG15-IFN-γ pathway cause increased susceptibility to intra-macrophagic pathogens, particularly to Mycobacteria. Therefore, the purpose of this study was to characterize the mutations in genes of the IL-12/23/ISG15-IFN-γ pathway in severe tuberculosis (TB) patients. METHODS Clinically suspected TB was initially confirmed in four patients (P) (P1, P2, P3, and P4) using the GeneXpert MTB/RIF and culturing techniques. The patients' Peripheral blood mononuclear cells (PBMCs) were then subjected to ELISA to measure Interleukin 12 (IL-12) and interferon gamma (IFN-γ). Flow cytometry was used to detect the surface expressions of IFN-γR1 and IFN-γR2 as well as IL-12Rβ1and IL-12Rβ2 on monocytes and T lymphocytes, respectively.The phosphorylation of signal transducer and activator of transcription 1(STAT1) on monocytes and STAT4 on T lymphocytes were also detected by flow cytometry. Sanger sequencing was used to identify mutations in the IL-12Rβ1, STAT1, NEMO, and CYBB genes. RESULTS P1's PBMCs exhibited reduced IFN-γ production, while P2's and P3's PBMCs exhibited impaired IL-12 induction. Low IL-12Rβ1 surface expression and reduced STAT4 phosphorylation were demonstrated by P1's T lymphocytes, while impaired STAT1 phosphorylation was detected in P2's monocytes. The impaired IκB-α degradation and abolished H2O2 production in monocytes and neutrophils of P3 and P4 were observed, respectively. Sanger sequencing revealed novel nonsense homozygous mutation: c.191 G>A/p.W64 * in exon 3 of the IL-12Rβ1 gene in P1, novel missense homozygous mutation: c.107 A>T/p.Q36L in exon 3 of the STAT1 gene in P2, missense hemizygous mutation:: c.950 A>C/p.Q317P in exon 8 of the NEMO gene in P3, and nonsense hemizygous mutation: c.868 C>T/p.R290X in exon 8 of CYBB gene in P4. CONCLUSION Our findings broaden the clinical and genetic spectra associated with IL-12/23/ISG15-IFN-γ axis anomalies. Additionally, our data suggest that TB patients in Pakistan should be investigated for potential genetic defects due to high prevalence of parental consanguinity and increased incidence of TB in the country.
Collapse
Affiliation(s)
- Sajjad Ahmad
- Institute of Basic Medical Science, Khyber Medical University, Peshawar, KP, Pakistan
| | - Jawad Ahmed
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Eman H Khalifa
- Al Baha University Faculty of Applied Medical Sciences, Saudi Arabia
| | - Farhad Ali Khattak
- Research & development Cell, Khyber College of Dentistry (KCD), Peshawar, Pakistan
| | - Anwar Sheed Khan
- Provincial TB Reference laboratory, Hayatabad Medical Complex, Peshawar, PK, Pakistan
| | - Syed Umar Farooq
- Department of oral pathology, Khyber College of Dentistry, Peshawar. Pakistan
| | | | | | - Nadeem Ullah
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden.
| | - Taj Ali Khan
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan; Division of Infectious Diseases & Global Medicine,Department of Medicine,University of Florida, Gainesville, FL,United States.
| |
Collapse
|
4
|
Wahl SM. Howard A. Young's 4 Decades in Science: More Than Just Experiments. J Interferon Cytokine Res 2022; 42:611-617. [PMID: 35944271 PMCID: PMC9835286 DOI: 10.1089/jir.2022.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 04/24/2022] [Indexed: 01/21/2023] Open
Affiliation(s)
- Sharon M. Wahl
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Kilinç G, Saris A, Ottenhoff THM, Haks MC. Host-directed therapy to combat mycobacterial infections. Immunol Rev 2021; 301:62-83. [PMID: 33565103 PMCID: PMC8248113 DOI: 10.1111/imr.12951] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/27/2020] [Indexed: 12/27/2022]
Abstract
Upon infection, mycobacteria, such as Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM), are recognized by host innate immune cells, triggering a series of intracellular processes that promote mycobacterial killing. Mycobacteria, however, have developed multiple counter‐strategies to persist and survive inside host cells. By manipulating host effector mechanisms, including phagosome maturation, vacuolar escape, autophagy, antigen presentation, and metabolic pathways, pathogenic mycobacteria are able to establish long‐lasting infection. Counteracting these mycobacteria‐induced host modifying mechanisms can be accomplished by host‐directed therapeutic (HDT) strategies. HDTs offer several major advantages compared to conventional antibiotics: (a) HDTs can be effective against both drug‐resistant and drug‐susceptible bacteria, as well as potentially dormant mycobacteria; (b) HDTs are less likely to induce bacterial drug resistance; and (c) HDTs could synergize with, or shorten antibiotic treatment by targeting different pathways. In this review, we will explore host‐pathogen interactions that have been identified for Mtb for which potential HDTs impacting both innate and adaptive immunity are available, and outline those worthy of future research. We will also discuss possibilities to target NTM infection by HDT, although current knowledge regarding host‐pathogen interactions for NTM is limited compared to Mtb. Finally, we speculate that combinatorial HDT strategies can potentially synergize to achieve optimal mycobacterial host immune control.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Ihedioha O, Potter AA, Chen JM. Poor stimulation of bovine dendritic cells by Mycobacterium bovis culture supernatant and surface extract is associated with decreased activation of ERK and NF- κB and higher expression of SOCS1 and 3. Innate Immun 2020; 26:537-546. [PMID: 32513050 PMCID: PMC7491241 DOI: 10.1177/1753425920929759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cell envelope of pathogenic mycobacteria interfaces with the host. As such, the interaction of bacterial products localized at or released from the cell surface with the host’s immune system can determine the fate of the bacterium in its host. In this study, the effects of three different types of Mycobacterium bovis cell envelope fractions—purified protein derivative, total cell wall lipids and culture supernatant and surface extract—on bovine dendritic cells were assessed. We found that the culture supernatant and surface extract fraction induced little to no production of the pro-inflammatory cytokines TNF-α and IL-12 in bovine dendritic cells. Moreover, this muted response was associated with poor activation of ERK and NF-κB, both of which are critical for the pro-inflammatory response. Furthermore, culture supernatant and surface extract treatment increased the expression of suppressor of cytokine signaling 1 and 3, both of which are negative regulators of pro-inflammatory signaling, in bovine dendritic cells. These observations taken together suggest the M. bovis culture supernatant and surface extract fraction contain immunomodulatory molecules that may aid in M. bovis pathogenesis.
Collapse
Affiliation(s)
- Olivia Ihedioha
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Canada.,Vaccine and Infectious Disease Organization, International Vaccine Centre, University of Saskatchewan, Canada
| | - Andrew A Potter
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Canada.,Vaccine and Infectious Disease Organization, International Vaccine Centre, University of Saskatchewan, Canada
| | - Jeffrey M Chen
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Canada.,Vaccine and Infectious Disease Organization, International Vaccine Centre, University of Saskatchewan, Canada
| |
Collapse
|
7
|
Convery O, Gargan S, Kickham M, Schroder M, O'Farrelly C, Stevenson NJ. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3. FASEB J 2019; 33:8732-8744. [PMID: 31163989 DOI: 10.1096/fj.201800629rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viruses use a spectrum of immune evasion strategies that enable infection and replication. The acute phase of hepatitis C virus (HCV) infection is characterized by nonspecific and often mild clinical symptoms, suggesting an immunosuppressive mechanism that, unless symptomatic liver disease presents, allows the virus to remain largely undetected. We previously reported that HCV induced the regulatory protein suppressor of cytokine signaling (SOCS)3, which inhibited TNF-α-mediated inflammatory responses. However, the mechanism by which HCV up-regulates SOCS3 remains unknown. Here we show that the HCV protein, p7, enhances both SOCS3 mRNA and protein expression. A p7 inhibitor reduced SOCS3 induction, indicating that p7's ion channel activity was required for optimal up-regulation of SOCS3. Short hairpin RNA and chemical inhibition revealed that both the Janus kinase-signal transducer and activator of transcription (JAK-STAT) and MAPK pathways were required for p7-mediated induction of SOCS3. HCV-p7 expression suppressed TNF-α-mediated IκB-α degradation and subsequent NF-κB promoter activity, revealing a new and functional, anti-inflammatory effect of p7. Together, these findings identify a molecular mechanism by which HCV-p7 induces SOCS3 through STAT3 and ERK activation and demonstrate that p7 suppresses proinflammatory responses to TNF-α, possibly explaining the lack of inflammatory symptoms observed during early HCV infection.-Convery, O., Gargan, S., Kickham, M., Schroder, M., O'Farrelly, C., Stevenson, N. J. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3.
Collapse
Affiliation(s)
- Orla Convery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Mizuno S, Soma S, Inada H, Kanuma T, Matsuo K, Yasutomi Y. SOCS1 Antagonist–Expressing Recombinant Bacillus Calmette–Guérin Enhances Antituberculosis Protection in a Mouse Model. THE JOURNAL OF IMMUNOLOGY 2019; 203:188-197. [DOI: 10.4049/jimmunol.1800694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 04/22/2019] [Indexed: 11/19/2022]
|
9
|
Abate G, Hamzabegovic F, Eickhoff CS, Hoft DF. BCG Vaccination Induces M. avium and M. abscessus Cross-Protective Immunity. Front Immunol 2019; 10:234. [PMID: 30837992 PMCID: PMC6389677 DOI: 10.3389/fimmu.2019.00234] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 01/28/2019] [Indexed: 01/14/2023] Open
Abstract
Pulmonary non-tuberculous mycobacterial (NTM) infections particularly caused by Mycobacterium avium complex (MAC) and Mycobacterium abscessus (MAB) are becoming major health problems in the U.S. New therapies or vaccines which will help prevent the disease, shorten treatment duration and/or increase treatment success rates are urgently needed. This study was conducted with the objective of testing the hypothesis that Bacillus Calmette Guerin (BCG), a vaccine used for prevention of serious forms of tuberculosis (TB) in children and adolescents in tuberculosis hyperendemic countries, induces cross-protective T cell immunity against Mycobacterium avium (MAV) and MAB. Human TB and NTM cross-protective T cells were quantified using flow cytometric assays. The ability of BCG expanded T cells to inhibit the intracellular growth of MAV and MAB was assessed in co-cultures with infected autologous macrophages. In both BCG-vaccinated and M. tuberculosis (Mtb)-infected mice, NTM cross-reactive immunity was measured using IFN-γ ELISPOT assays. Our results demonstrate the following key findings: (i) peripheral blood mononuclear cells from TB skin test-positive individuals contain MAV and MAB cross-reactive T cells, (ii) both BCG vaccination and Mtb infection of mice induce MAV and MAB cross-reactive splenic cells, (iii) BCG-expanded T cells inhibit intracellular MAV and MAB, (iv) CD4, CD8, and γδ T cells play important roles in inhibition of intracellular MAV and MAB and (v) BCG vaccination of healthy volunteers induces TB and NTM cross-reactive T cells. In conclusion, BCG-vaccination induces NTM cross-reactive immunity, and has the potential for use as a vaccine or immunotherapy to prevent and/or treat pulmonary NTM disease.
Collapse
Affiliation(s)
- Getahun Abate
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University, St. Louis, MO, United States,*Correspondence: Getahun Abate
| | - Fahreta Hamzabegovic
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University, St. Louis, MO, United States
| | - Christopher S. Eickhoff
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University, St. Louis, MO, United States
| | - Daniel F. Hoft
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University, St. Louis, MO, United States,Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
10
|
Primary Role of Suppressor of Cytokine Signaling 1 in Mycobacterium bovis BCG Infection. Infect Immun 2018; 86:IAI.00376-18. [PMID: 30181351 DOI: 10.1128/iai.00376-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/29/2018] [Indexed: 11/20/2022] Open
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a negative regulator of JAK/STAT signaling and is induced by mycobacterial infection. To understand the major function of SOCS1 during infection, we established a novel system in which recombinant Mycobacterium bovis bacillus Calmette-Guérin expressed dominant-negative SOCS1 (rBCG-SOCS1DN) because it would not affect the function of SOCS1 in uninfected cells. When C57BL/6 mice and RAG1-/- mice were intratracheally inoculated with rBCG-SOCS1DN, the amount of rBCG-SOCS1DN in the lungs was significantly reduced compared to that in the lungs of mice inoculated with a vector control counterpart and wild-type BCG. However, these significant differences were not observed in NOS2-/- mice and RAG1-/- NOS2-/- double-knockout mice. These findings demonstrated that SOCS1 inhibits nitric oxide (NO) production to establish mycobacterial infection and that rBCG-SOCS1DN has the potential to be a powerful tool for studying the primary function of SOCS1 in mycobacterial infection.
Collapse
|
11
|
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
|
12
|
Marino R, Capoferri R, Panelli S, Minozzi G, Strozzi F, Trevisi E, Snel GGM, Ajmone-Marsan P, Williams JL. Johne's disease in cattle: an in vitro model to study early response to infection of Mycobacterium avium subsp. paratuberculosis using RNA-seq. Mol Immunol 2017; 91:259-271. [PMID: 28988040 DOI: 10.1016/j.molimm.2017.08.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/07/2017] [Accepted: 08/21/2017] [Indexed: 02/07/2023]
Abstract
Johne's disease is a chronic granulomatous enteritis caused by Mycobacterium avium subsp. paratubercolosis (MAP) which affects ruminants worldwide and has a significant economic impact. MAP has also been associated with human Crohn's disease, although this connection is not well established. MAP is highly adapted for survival within host macrophages and prevents macrophage activation, blocks phagosome acidification and maturation, and attenuates presentation of antigens to the immune system. The consequence is a very long silent infection before clinical signs are observed. The present work examined the transcriptome of bovine monocyte-derived macrophages (MDM) infected with the L1 strain of MAP at 2h, 6h and 24h post infection using RNA-seq. Pathway over-representation analysis of genes differentially expressed between infected vs. control MDM identified that immune related pathways were affected. Genes belonging to the cytokine-cytokine receptor interaction pathway and members of the JAK-STAT pathway, which is involved in the regulation of immune response, were up-regulated. However, in parallel inhibitors of immune functions were activated, including suppressor of cytokine signaling (SOCS) and cytokine-inducible SH2-containing protein (CISH), which most likely suppresses IFNγ and the JAK/STAT signaling cascade in infected MDM, which may favour MAP survival. After exposure, macrophages phagocytise pathogens, activate the complement cascade and the adaptive immune system through the antigen presentation process. However, data presented here suggest that genes related to phagocytosis and lysosome function are down regulated in MAP infected MDM. Genes of MHC class II and complement pathway were also down-regulated. This study therefore shows that MAP infection is associated with changes in expression of genes related to the host immune response that may affect its ability to survive and multiply inside the host cell.
Collapse
Affiliation(s)
- Rosanna Marino
- CREA Research Centre for Animal Production and Aquaculture, Via Antonio Lombardo 11, 26900 Lodi, Italy; Istituto Sperimentale Italiano "Lazzaro Spallanzani", 26027, Rivolta d'Adda, Cremona, Italy; Institute of Zootechnics, Università Cattolica del S. Cuore, Via Emilia Parmense 84, 29122 Piacenza, Italy.
| | - Rossana Capoferri
- Istituto Sperimentale Italiano "Lazzaro Spallanzani", 26027, Rivolta d'Adda, Cremona, Italy.
| | - Simona Panelli
- Parco Tecnologico Padano, via Einstein, 26900 Lodi, Italy.
| | | | | | - Erminio Trevisi
- Institute of Zootechnics, Università Cattolica del S. Cuore, Via Emilia Parmense 84, 29122 Piacenza, Italy; Nutrigenomics and Proteomic Research Center - PRONUTRIGEN, Università Cattolica del S. Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy.
| | | | - Paolo Ajmone-Marsan
- Institute of Zootechnics, Università Cattolica del S. Cuore, Via Emilia Parmense 84, 29122 Piacenza, Italy; Nutrigenomics and Proteomic Research Center - PRONUTRIGEN, Università Cattolica del S. Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy.
| | - John L Williams
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA 5371, Australia.
| |
Collapse
|
13
|
Schulte JN, Brockmann GA, Kreuzer-Redmer S. Feeding a high dosage of zinc oxide affects suppressor of cytokine gene expression in Salmonella Typhimurium infected piglets. Vet Immunol Immunopathol 2016; 178:10-3. [DOI: 10.1016/j.vetimm.2016.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 01/31/2023]
|
14
|
Surolia R, Karki S, Wang Z, Kulkarni T, Li FJ, Vohra S, Batra H, Nick JA, Duncan SR, Thannickal VJ, Steyn AJC, Agarwal A, Antony VB. Attenuated heme oxygenase-1 responses predispose the elderly to pulmonary nontuberculous mycobacterial infections. Am J Physiol Lung Cell Mol Physiol 2016; 311:L928-L940. [PMID: 27694475 DOI: 10.1152/ajplung.00397.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 09/22/2016] [Indexed: 12/22/2022] Open
Abstract
Pulmonary infections with nontuberculous mycobacteria (P-NTM), such as by Mycobacterium avium complex (M. avium), are increasingly found in the elderly, but the underlying mechanisms are unclear. Recent studies suggest that adaptive immunity is necessary, but not sufficient, for host defense against mycobacteria. Heme oxygenase-1 (HO-1) has been recognized as a critical modulator of granuloma formation and programmed cell death in mycobacterial infections. Old mice (18-21 mo) infected with M. avium had attenuated HO-1 response with diffuse inflammation, high burden of mycobacteria, poor granuloma formation, and decreased survival (45%), while young mice (4-6 mo) showed tight, well-defined granuloma, increased HO-1 expression, and increased survival (95%). To further test the role of HO-1 in increased susceptibility to P-NTM infections in the elderly, we used old and young HO-1+/+ and HO-1-/- mice. The transcriptional modulation of the JAK/STAT signaling pathway in HO-1-/- mice due to M. avium infection demonstrated similarities to infected wild-type old mice with upregulation of SOCS3 and inhibition of Bcl2. Higher expression of SOCS3 with downregulation of Bcl2 resulted in higher macrophage death via cellular necrosis. Finally, peripheral blood monocytes (PBMCs) from elderly patients with P-NTM also demonstrated attenuated HO-1 responses after M. avium stimulation and increased cell death due to cellular necrosis (9.69% ± 2.02) compared with apoptosis (4.75% ± 0.98). The augmented risk for P-NTM in the elderly is due, in part, to attenuated HO-1 responses, subsequent upregulation of SOCS3, and inhibition of Bcl2, leading to programmed cell death of macrophages, and sustained infection.
Collapse
Affiliation(s)
- Ranu Surolia
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Suman Karki
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zheng Wang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tejaswini Kulkarni
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Fu Jun Li
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shikhar Vohra
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hitesh Batra
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, University of Colorado, Denver, Colorado
| | - Steven R Duncan
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor J Thannickal
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama
| | - Adrie J C Steyn
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama.,KwaZulu-Natal Research Institute for TB and HIV, Durban, South Africa; and
| | - Anupam Agarwal
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama.,Birmingham VA Medical Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Veena B Antony
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama;
| |
Collapse
|
15
|
Titanium dioxide nanoparticles augment allergic airway inflammation and Socs3 expression via NF-κB pathway in murine model of asthma. Biomaterials 2016; 92:90-102. [PMID: 27057692 DOI: 10.1016/j.biomaterials.2016.03.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 03/05/2016] [Accepted: 03/10/2016] [Indexed: 12/25/2022]
Abstract
Titanium dioxide nanoparticles (nTiO2) previously considered to possess relatively low toxicity both in vitro and in vivo, although classified as possibly carcinogenic to humans. Also, their adjuvant potential has been reported to promote allergic sensitization and modulate immune responses. Previously, in OVA induced mouse model of asthma we found high expression of Socs3 and low expression of Stat3 and IL-6. However, a clear understanding regarding the signaling pathways associated with nTiO2 adjuvant effect in mouse model of asthma is lacking. In the present study we investigated the status of Stat3/IL-6 and Socs3 and their relationship with NF-κB, with nTiO2 as an adjuvant in mouse model of asthma. nTiO2 when administered with ovalbumin (OVA) during sensitization phase augmented airway hyper-responsiveness (AHR), biochemical markers of lung damage and a mixed Th2/Th1 dependent immune response. At the same time, we observed significant elevation in the levels of Stat3, Socs3, NF-κB, IL-6 and TNF-α. Furthermore, transient in vivo blocking of NF-κB by NF-κB p65 siRNA, downregulated the expression of Socs3, IL-6 and TNF-α. Our study, thus, shows that nTiO2 exacerbate the inflammatory responses in lungs of pre-sensitized allergic individuals and that these changes are regulated via NF-κB pathway.
Collapse
|
16
|
Schistosoma mansoni Soluble Egg Antigens Induce Expression of the Negative Regulators SOCS1 and SHP1 in Human Dendritic Cells via Interaction with the Mannose Receptor. PLoS One 2015; 10:e0124089. [PMID: 25897665 PMCID: PMC4405200 DOI: 10.1371/journal.pone.0124089] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/25/2015] [Indexed: 12/31/2022] Open
Abstract
Schistosomiasis is a common debilitating human parasitic disease in (sub)tropical areas, however, schistosome infections can also protect against a variety of inflammatory diseases. This has raised broad interest in the mechanisms by which Schistosoma modulate the immune system into an anti-inflammatory and regulatory state. Human dendritic cells (DCs) show many phenotypic changes upon contact with Schistosoma mansoni soluble egg antigens (SEA). We here show that oxidation of SEA glycans, but not heat-denaturation, abrogates the capacity of SEA to suppress both LPS-induced cytokine secretion and DC proliferation, indicating an important role of SEA glycans in these processes. Remarkably, interaction of SEA glycans with DCs results in a strongly increased expression of Suppressor Of Cytokine Signalling1 (SOCS1) and SH2-containing protein tyrosine Phosphatase-1 (SHP1), important negative regulators of TLR4 signalling. In addition, SEA induces the secretion of transforming growth factor β (TGF-β), and the surface expression of the costimulatory molecules Programmed Death Ligand-1 (PD-L1) and OX40 ligand (OX40L), which are known phenotypic markers for the capacity of DCs to polarize naïve T cells into Th2/Treg cell subsets. Inhibition of mannose receptor (MR)-mediated internalization of SEA into DCs by blocking with allyl α-D-mannoside or anti-MR antibodies, significantly reduced SOCS1 and SHP1 expression. In conclusion, we demonstrate that SEA glycans are essential for induction of enhanced SOCS1 and SHP1 levels in DCs via the MR. Our data provide novel mechanistic evidence for the potential of S. mansoni SEA glycans to modulate human DCs, which may contribute to the capacity of SEA to down-regulate inflammatory responses.
Collapse
|
17
|
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]
|
18
|
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
|
19
|
Zhao Q, Li W, Chen T, He Y, Deng W, Luo H, Xie J. Mycobacterium tuberculosis serine protease Rv3668c can manipulate the host-pathogen interaction via Erk-NF-κB axis-mediated cytokine differential expression. J Interferon Cytokine Res 2014; 34:686-98. [PMID: 24684623 DOI: 10.1089/jir.2013.0071] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Tuberculosis caused by Mycobacterium tuberculosis (MTB) remains a serious global public health concern. About one-third of the global population has been latently infected with this pathogen. MTB proteases are important virulence factors and involve in subverting the host immunity. MTB protease Rv3668c was implicated in drug action and dormancy by Gene Expression Omnibus data. To define the role of Rv3668c in pathogen-host interaction, we constructed recombinant strain Mycobacterium smegmatis-Rv3668c (Ms-Rv3668c). The resultant strains were used to challenge the human macrophage cell line U937. The cytokine levels and the survival of recombinants and macrophages were monitored. The results showed that recombinant Ms-Rv3668c specifically upregulated the secretion of proinflammatory cytokines TNF-α, IL-1β, and IL-6 and downregulated the secretion of anti-inflammatory cytokine IL-10 by U937 cells, consistent with the upregulated transcription of TNF-α and IL-1β. Rv3668c recombinants demonstrated prolonged survival within the U937 cells and accelerated the death of the host cells. Inhibitor experiments showed that the ERK-NF-κB axis was involved in the Rv3668c-triggered TNF-α and IL-1β changes. These results provided evidence for the engagement of Rv3668c in the interaction between Mycobacterium and host.
Collapse
Affiliation(s)
- Quanju Zhao
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, and Key Laboratory of Ministry of Education and Eco-Environment of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University , Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Corti M, Palmero D. Mycobacterium aviumcomplex infection in HIV/AIDS patients. Expert Rev Anti Infect Ther 2014; 6:351-63. [PMID: 18588499 DOI: 10.1586/14787210.6.3.351] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Marcelo Corti
- HIV/AIDS Division, Infectious Diseases FJ Muñiz Hospital, Buenos Aires, Argentina.
| | | |
Collapse
|
21
|
Holla S, Kurowska-Stolarska M, Bayry J, Balaji KN. Selective inhibition of IFNG-induced autophagy by Mir155- and Mir31-responsive WNT5A and SHH signaling. Autophagy 2013; 10:311-30. [PMID: 24343269 DOI: 10.4161/auto.27225] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Autophagy is one of the major immune mechanisms engaged to clear intracellular infectious agents. However, several pathogens have evolved strategies to evade autophagy. Here, we demonstrated that Mycobacteria, Shigella, and Listeria but not Klebsiella, Staphylococcus, and Escherichia inhibit IFNG-induced autophagy in macrophages by evoking selective and robust activation of WNT and SHH pathways via MTOR. Utilization of gain- or loss-of-function analyses as well as mir155-null macrophages emphasized the role of MTOR-responsive epigenetic modifications in the induction of Mir155 and Mir31. Importantly, cellular levels of PP2A, a phosphatase, were regulated by Mir155 and Mir31 to fine-tune autophagy. Diminished expression of PP2A led to inhibition of GSK3B, thus facilitating the prolonged activation of WNT and SHH signaling pathways. Sustained WNT and SHH signaling effectuated the expression of anti-inflammatory lipoxygenases, which in tandem inhibited IFNG-induced JAK-STAT signaling and contributed to evasion of autophagy. Altogether, these results established a role for new host factors and inhibitory mechanisms employed by the pathogens to limit autophagy, which could be targeted for therapeutic interventions.
Collapse
Affiliation(s)
- Sahana Holla
- Department of Microbiology and Cell Biology; Indian Institute of Science; Bangalore, Karnataka India
| | | | - Jagadeesh Bayry
- Centre de Recherche des Cordeliers; Institut National de la Santé et de la Recherche Médicale; Paris, France
| | | |
Collapse
|
22
|
Delgado-Ortega M, Marc D, Dupont J, Trapp S, Berri M, Meurens F. SOCS proteins in infectious diseases of mammals. Vet Immunol Immunopathol 2012; 151:1-19. [PMID: 23219158 PMCID: PMC7112700 DOI: 10.1016/j.vetimm.2012.11.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 10/31/2012] [Accepted: 11/13/2012] [Indexed: 12/17/2022]
Abstract
As for most biological processes, the immune response to microbial infections has to be tightly controlled to remain beneficial for the host. Inflammation is one of the major consequences of the host's immune response. For its orchestration, this process requires a fine-tuned interplay between interleukins, endothelial cells and various types of recruited immune cells. Suppressors of cytokine signalling (SOCS) proteins are crucially involved in the complex control of the inflammatory response through their actions on various signalling pathways including the JAK/STAT and NF-κB pathways. Due to their cytokine regulatory functions, they are frequent targets for exploitation by infectious agents trying to escape the host's immune response. This review article aims to summarize our current knowledge regarding SOCS family members in the different mammalian species studied so far, and to display their complex molecular interactions with microbial pathogens.
Collapse
Affiliation(s)
- Mario Delgado-Ortega
- Institut National de la Recherche Agronomique (INRA), UMR1282 Infectiologie et Santé Publique, F-37380 Nouzilly, France
| | | | | | | | | | | |
Collapse
|
23
|
Vázquez N, Rekka S, Gliozzi M, Feng CG, Amarnath S, Orenstein JM, Wahl SM. Modulation of innate host factors by Mycobacterium avium complex in human macrophages includes interleukin 17. J Infect Dis 2012; 206:1206-17. [PMID: 22930805 DOI: 10.1093/infdis/jis492] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Although opportunistic infections due to Mycobacterium avium complex (MAC) have been less common since the introduction of highly active antiretroviral therapy, globally, human immunodeficiency virus-1 (HIV-1)-positive patients remain predisposed to these infections. Absence of a properly functioning acquired immune response allows MAC persistence within macrophages localized in lymph nodes coinfected with HIV and MAC. Although a deficiency in interferon γ appears to play a part in the ability of MAC to deflect the macrophage-associated antimicrobial attack, questions about this process remain. Our study examines the ability of MAC to regulate interleukin 17 (IL-17), a proinflammatory cytokine involved in host cell recruitment. METHODS Coinfected lymph nodes were examined for IL-17 by immunohistochemical analysis. In vitro, macrophages exposed to mycobacteria were evaluated for transcription activities, proteins, and signaling pathways responsible for IL-17 expression. Infected macrophages were also analyzed for expression of interleukin 21 (IL-21) and negative regulators of immune responses. RESULTS Infection of macrophages triggered synthesis of IL-17, correlating with IL-17 expression by macrophages in coinfected lymph nodes. Infected macrophages exposed to exogenous IL-17 expressed CXCL10, which favors recruitment of new macrophages as targets for infection. Blockade of nuclear factor κ-light-chain-enhancer of activated B cells and mitogen-activated protein kinase pathways suppressed mycobacteria-induced IL-17 expression. MAC triggered expression of IL-21, IRF4, and STAT3 genes related to IL-17 regulation, as well as expression of the negative immunoregulators CD274(PD-L1) and suppressors of cytokine signaling. CONCLUSIONS MAC-infected macrophages can provide an alternative source for IL-17 that favors accumulation of new targets for perpetuating bacterial and viral infection while suppressing host antimicrobial immune responses.
Collapse
Affiliation(s)
- Nancy Vázquez
- Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, Bethesda, MD 20892-4352, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Demirel I, Säve S, Kruse R, Persson K. Expression of suppressor of cytokine signalling 3 (SOCS3) in human bladder epithelial cells infected with uropathogenic Escherichia coli. APMIS 2012; 121:158-67. [PMID: 23030674 DOI: 10.1111/j.1600-0463.2012.02951.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/27/2012] [Indexed: 11/29/2022]
Abstract
Suppressor of cytokine signalling (SOCS) proteins inhibit pro-inflammatory signalling mediated by Janus-activated kinase (JAK)-signal transducer and activator of transcription (STAT) pathways. To evade the immune response some pathogens appear to modify the host SOCS proteins. Uropathogenic Escherichia coli (UPEC) are able to subvert the host response evoked by bladder epithelial cells, but the mechanisms are not fully understood. The objective of this study was to investigate whether UPEC can modify the host SOCS and STAT3 response. Real time RT-PCR studies demonstrated an increased SOCS1 and SOCS3 expression in the isolated human bladder epithelial cell lines (RT-4 and 5637) in response to cytokines. UPEC strain IA2 increased SOCS3, but not SOCS1, mRNA levels with a peak at 6 h after infection. The increase of SOCS3 was confirmed at the protein level by Western blotting. The UPEC strain IA2 caused a time-dependent decrease in the phosphorylation of STAT3. This study demonstrates that UPEC are able to affect SOCS3 and STAT3 signalling in human uroepithelial cells. The finding that UPEC are able to induce mediators involved in suppression of host cytokine signalling may help to elucidate how UPEC may circumvent the host response during urinary tract infection.
Collapse
Affiliation(s)
- Isak Demirel
- Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, Sweden.
| | | | | | | |
Collapse
|
25
|
Wenink MH, Santegoets KCM, Butcher J, van Bon L, Lamers-Karnebeek FGM, van den Berg WB, van Riel PLCM, McInnes IB, Radstake TRDJ. Impaired dendritic cell proinflammatory cytokine production in psoriatic arthritis. ACTA ACUST UNITED AC 2011; 63:3313-22. [DOI: 10.1002/art.30577] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Stutz A, Kessler H, Kaschel ME, Meissner M, Dalpke AH. Cell invasion and strain dependent induction of suppressor of cytokine signaling-1 by Toxoplasma gondii. Immunobiology 2011; 217:28-36. [PMID: 22015046 DOI: 10.1016/j.imbio.2011.08.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 08/17/2011] [Accepted: 08/21/2011] [Indexed: 01/12/2023]
Abstract
Toxoplasma gondii is an intracellular parasite that has to cope with the microbicidal actions of IFNγ. Previously we reported that parasite-mediated induction of suppressor of cytokine signaling protein 1 (SOCS1) contributes to inhibition of IFNγ signaling. However, the signaling requirements remained elusive. We now show that induction of SOCS1 and inhibition of nitric oxide production by IFNγ was independent of stimulation of Toll-like receptors. Instead, infection by T. gondii resulted in induction of egr transcription factors which have been reported to regulate SOCS expression. Indeed, induction of egr2 as well as SOCS1 was dependent on p38 MAP kinase and blockade of egr inhibited SOCS1 expression. Moreover, we found that Mic8, a previously identified invasion factor of T. gondii, was necessary for SOCS1 regulation and escape of IFNγ mediated nitric oxide secretion within macrophages. Surprisingly, when further analyzing Mic8 deficient parasites we noted that inhibition of IFNγ mediated up-regulation of MHC-class II and ICAM1 molecules was independent of cell invasion. Furthermore, these inhibitory effects were equally observed in type I and II strains of T. gondii and were dependent on excreted and secreted antigens. In contrast, only the virulent RH type I strain additionally induced SOCS1 and efficiently inhibited nitric oxide secretion by IFNγ. The results show that T. gondii makes use of two different mechanisms to escape from IFNγ activity with one mode being strain dependent and relying on active cell invasion and SOCS1 induction.
Collapse
Affiliation(s)
- Andrea Stutz
- Dept. of Infectious Diseases - Medical Microbiology and Hygiene, University Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
27
|
Amaral EP, Kipnis TL, de Carvalho ECQ, da Silva WD, Leão SC, Lasunskaia EB. Difference in virulence of Mycobacterium avium isolates sharing indistinguishable DNA fingerprint determined in murine model of lung infection. PLoS One 2011; 6:e21673. [PMID: 21738761 PMCID: PMC3125297 DOI: 10.1371/journal.pone.0021673] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 06/07/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Opportunistic Mycobacterium avium typically causes disease in immunocompromised patients and in some groups of apparently healthy individuals. The high virulence of some bacterial lineages increases the disease risk. High-resolution molecular genotyping studies of M. avium clinical isolates demonstrated that some genotype patterns were more prevalent than others, suggesting that close genetic relatedness of these successful isolates sharing a similar genotype could determine similar biological properties associated with high virulence. METHODS AND FINDINGS In this study, we aimed to compare the virulence and pathogenic properties of two epidemiologically unrelated M. avium isolates sharing an indistinguishable DNA fingerprint in a well-characterized model of pulmonary infection in mice, resistant or susceptible to mycobacteria. The mice, C57BL/6 wild- type or IFN-gamma gene disrupted (GKO), respectively, were intratracheally infected with two isolates, H27 (human blood isolate) and P104 (pig lymph node isolate), and the lungs were examined for bacterial loads, histopathology and cytokine gene expression. The obtained data demonstrated significant differences in the virulence properties of these strains. Although the H27 strain grew significantly faster than P104 in the early stage of infection, this bacterium induced protective immunity that started to reduce bacterial numbers in the wild-type mice, whereas the P104 strain established a chronic infection. In the GKO mice, both strains were capable of causing a chronic infection, associated with higher bacterial burdens and severe lung pathology, in a similar manner. CONCLUSIONS/SIGNIFICANCE The results demonstrated that the studied isolates differed in the pathogenic properties although were indistinguishable by actually widely used genotyping techniques demonstrating that the genotype similarity does not predict similarity in virulence of M. avium isolates.
Collapse
Affiliation(s)
- Eduardo Pinheiro Amaral
- Laboratory of Biology of Recognition, Universidade Estadual do Norte Fluminense, Campos, Rio de Janeiro, Brazil
| | - Thereza Liberman Kipnis
- Laboratory of Biology of Recognition, Universidade Estadual do Norte Fluminense, Campos, Rio de Janeiro, Brazil
| | | | | | - Sylvia Cardoso Leão
- Department of Microbiology, Immunology and Parasitology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Elena B. Lasunskaia
- Laboratory of Biology of Recognition, Universidade Estadual do Norte Fluminense, Campos, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Carow B, Ye XQ, Gavier-Widén D, Bhuju S, Oehlmann W, Singh M, Sköld M, Ignatowicz L, Yoshimura A, Wigzell H, Rottenberg ME. Silencing suppressor of cytokine signaling-1 (SOCS1) in macrophages improves Mycobacterium tuberculosis control in an interferon-gamma (IFN-gamma)-dependent manner. J Biol Chem 2011; 286:26873-87. [PMID: 21622562 DOI: 10.1074/jbc.m111.238287] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Protection against infection with Mycobacterium tuberculosis demands IFN-γ. SOCS1 has been shown to inhibit responses to IFN-γ and might thereby play a central role in the outcome of infection. We found that M. tuberculosis is a highly efficient stimulator of SOCS1 expression in murine and human macrophages and in tissues from infected mice. Surprisingly, SOCS1 reduced responses to IL-12, resulting in an impaired IFN-γ secretion by macrophages that in turn accounted for a deteriorated intracellular mycobacterial control. Despite SOCS1 expression, mycobacteria-infected macrophages responded to exogenously added IFN-γ. SOCS1 attenuated the expression of the majority of genes modulated by M. tuberculosis infection of macrophages. Using a conditional knockdown strategy in mice, we found that SOCS1 expression by macrophages hampered M. tuberculosis clearance early after infection in vivo in an IFN-γ-dependent manner. On the other hand, at later time points, SOCS1 expression by non-macrophage cells protected the host from infection-induced detrimental inflammation.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 17177, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sharbati J, Lewin A, Kutz-Lohroff B, Kamal E, Einspanier R, Sharbati S. Integrated microRNA-mRNA-analysis of human monocyte derived macrophages upon Mycobacterium avium subsp. hominissuis infection. PLoS One 2011; 6:e20258. [PMID: 21629653 PMCID: PMC3101234 DOI: 10.1371/journal.pone.0020258] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 04/22/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Many efforts have been made to understand basal mechanisms of mycobacterial infections. Macrophages are the first line of host immune defence to encounter and eradicate mycobacteria. Pathogenic species have evolved different mechanisms to evade host response, e.g. by influencing macrophage apoptotic pathways. However, the underlying molecular regulation is not fully understood. A new layer of eukaryotic regulation of gene expression is constituted by microRNAs. Therefore, we present a comprehensive study for identification of these key regulators and their targets in the context of host macrophage response to mycobacterial infections. METHODOLOGY/PRINCIPAL FINDINGS We performed microRNA as well as mRNA expression analysis of human monocyte derived macrophages infected with several Mycobacterium avium hominissuis strains by means of microarrays as well as quantitative reverse transcription PCR (qRT-PCR). The data revealed the ability of all strains to inhibit apoptosis by transcriptional regulation of BCL2 family members. Accordingly, at 48 h after infection macrophages infected with all M. avium strains showed significantly decreased caspase 3 and 7 activities compared to the controls. Expression of let-7e, miR-29a and miR-886-5p were increased in response to mycobacterial infection at 48 h. The integrated analysis of microRNA and mRNA expression as well as target prediction pointed out regulative networks identifying caspase 3 and 7 as potential targets of let-7e and miR-29a, respectively. Consecutive reporter assays verified the regulation of caspase 3 and 7 by these microRNAs. CONCLUSIONS/SIGNIFICANCE We show for the first time that mycobacterial infection of human macrophages causes a specific microRNA response. We furthermore outlined a regulatory network of potential interactions between microRNAs and mRNAs. This study provides a theoretical concept for unveiling how distinct mycobacteria could manipulate host cell response. In addition, functional relevance was confirmed by uncovering the control of major caspases 3 and 7 by let-7e and miR-29a, respectively.
Collapse
Affiliation(s)
- Jutta Sharbati
- Institute of Veterinary Biochemistry, Freie Universitaet Berlin, Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
30
|
Nair S, Pandey AD, Mukhopadhyay S. The PPE18 protein of Mycobacterium tuberculosis inhibits NF-κB/rel-mediated proinflammatory cytokine production by upregulating and phosphorylating suppressor of cytokine signaling 3 protein. THE JOURNAL OF IMMUNOLOGY 2011; 186:5413-24. [PMID: 21451109 DOI: 10.4049/jimmunol.1000773] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mycobacterium tuberculosis bacteria are known to suppress proinflammatory cytokines like IL-12 and TNF-α for a biased Th2 response that favors a successful infection and its subsequent intracellular survival. However, the signaling pathways targeted by the bacilli to inhibit production of these cytokines are not fully understood. In this study, we demonstrate that the PPE18 protein of M. tuberculosis inhibits LPS-induced IL-12 and TNF-α production by blocking nuclear translocation of p50, p65 NF-κB, and c-rel transcription factors. We found that PPE18 upregulates the expression as well as tyrosine phosphorylation of suppressor of cytokine signaling 3 (SOCS3), and the phosphorylated SOCS3 physically interacts with IκBα-NF-κB/rel complex, inhibiting phosphorylation of IκBα at the serine 32/36 residues by IκB kinase-β, and thereby prevents nuclear translocation of the NF-κB/rel subunits in LPS-activated macrophages. Specific knockdown of SOCS3 by small interfering RNA enhanced IκBα phosphorylation, leading to increased nuclear levels of NF-κB/rel transcription factors vis-a-vis IL-12 p40 and TNF-α production in macrophages cotreated with PPE18 and LPS. The PPE18 protein did not affect the IκB kinase-β activity. Our study describes a novel mechanism by which phosphorylated SOCS3 inhibits NF-κB activation by masking the phosphorylation site of IκBα. Also, this study highlights the possible mechanisms by which the M. tuberculosis suppresses production of proinflammatory cytokines using PPE18.
Collapse
Affiliation(s)
- Shiny Nair
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad 500001, Andhra Pradesh, India
| | | | | |
Collapse
|
31
|
Comparative immunological and microbiological aspects of paratuberculosis as a model mycobacterial infection. Vet Immunol Immunopathol 2011; 148:29-47. [PMID: 21450348 DOI: 10.1016/j.vetimm.2011.03.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 02/12/2011] [Accepted: 03/03/2011] [Indexed: 11/20/2022]
Abstract
Paratuberculosis or Johne's disease of livestock, which is caused by Mycobacterium avium subsp. paratuberculosis (MAP), has increased in prevalence and expanded in geographic and host ranges over about 100 years. The slow and progressive spread of MAP reflects its substantial adaptation to its hosts, the technical limitations of diagnosis, the lack of practical therapeutic approaches, the lack of a vaccine that prevents transmission and the complexity and difficulty of the on-farm control strategies needed to prevent infection. More recently evidence has accumulated for an association of MAP with Crohn's disease in humans, adding to the pressure on animal health authorities to take precautions by controlling paratuberculosis. Mycobacterial infections invoke complex immune responses but the essential determinants of virulence and pathogenesis are far from clear. In this review we compare the features of major diseases in humans and animals that are caused by the pathogenic mycobacteria M. ulcerans, M. avium subsp. avium, M. leprae, M. tuberculosis and MAP. We seek to answer key questions: are the common mycobacterial infections of humans and animals useful "models" for each other, or are the differences between them too great to enable meaningful extrapolation? To simplify this, the immunopathogenesis of mycobacterial infections will be defined at cellular, tissue, animal and population levels and the key events at each level will be discussed. Many pathogenic processes are similar between divergent mycobacterial diseases, and at variance between virulent and avirulent isolates of mycobacteria, suggesting that the research on the pathogenesis of one mycobacterial disease will be informative for the others.
Collapse
|
32
|
Smith PD, Smythies LE, Shen R, Greenwell-Wild T, Gliozzi M, Wahl SM. Intestinal macrophages and response to microbial encroachment. Mucosal Immunol 2011; 4:31-42. [PMID: 20962772 PMCID: PMC3821935 DOI: 10.1038/mi.2010.66] [Citation(s) in RCA: 278] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Macrophages in the gastrointestinal mucosa represent the largest pool of tissue macrophages in the body. In order to maintain mucosal homeostasis, resident intestinal macrophages uniquely do not express the lipopolysaccharide (LPS) co-receptor CD14 or the IgA (CD89) and IgG (CD16, 32, and 64) receptors, yet prominently display Toll-like receptors (TLRs) 3-9. Remarkably, intestinal macrophages also do not produce proinflammatory cytokines in response to TLR ligands, likely because of extracellular matrix (stromal) transforming growth factor-β (TGF-β) dysregulation of nuclear factor (NF)-κB signal proteins and, via Smad signaling, expression of IκBα, thereby inhibiting NF-κB-mediated activities. Thus, in noninflamed mucosa, resident macrophages are inflammation anergic but retain avid scavenger and host defense function, an ideal profile for macrophages in close proximity to gut microbiota. In the event of impaired epithelial integrity during intestinal infection or inflammation, however, blood monocytes also accumulate in the lamina propria and actively pursue invading microorganisms through uptake and degradation of the organism and release of inflammatory mediators. Consequently, resident intestinal macrophages are inflammation adverse, but when the need arises, they receive assistance from newly recruited circulating monocytes.
Collapse
Affiliation(s)
- PD Smith
- Department of Medicine (Gastroenterology) University of Alabama at Birmingham Birmingham, Alabama 35294-2182, USA
| | - LE Smythies
- Department of Medicine (Gastroenterology) University of Alabama at Birmingham Birmingham, Alabama 35294-2182, USA
| | - R Shen
- Department of Medicine (Gastroenterology) University of Alabama at Birmingham Birmingham, Alabama 35294-2182, USA
| | - T Greenwell-Wild
- Oral Infection and Immunity Branch National Institute of Dental and Craniofacial Research National Institutes of Health Bethesda, MD 20892-4352, USA
| | - M Gliozzi
- Oral Infection and Immunity Branch National Institute of Dental and Craniofacial Research National Institutes of Health Bethesda, MD 20892-4352, USA
| | - SM Wahl
- Oral Infection and Immunity Branch National Institute of Dental and Craniofacial Research National Institutes of Health Bethesda, MD 20892-4352, USA
| |
Collapse
|
33
|
Mendoza-Coronel E, Camacho-Sandoval R, Bonifaz LC, López-Vidal Y. PD-L2 induction on dendritic cells exposed to Mycobacterium avium downregulates BCG-specific T cell response. Tuberculosis (Edinb) 2011; 91:36-46. [DOI: 10.1016/j.tube.2010.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 10/06/2010] [Accepted: 11/16/2010] [Indexed: 11/26/2022]
|
34
|
Sabbatini F, Bandera A, Ferrario G, Trabattoni D, Marchetti G, Franzetti F, Clerici M, Gori A. Qualitative immune modulation by interleukin-2 (IL-2) adjuvant therapy in immunological non responder HIV-infected patients. PLoS One 2010; 5:e14119. [PMID: 21124762 PMCID: PMC2993926 DOI: 10.1371/journal.pone.0014119] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 11/02/2010] [Indexed: 11/21/2022] Open
Abstract
Background Treatment of HIV-infected patients with interleukin-2 (IL-2) produces significant increases in CD4 T cell counts; however an associated qualitative improvement in cells function has yet to be conclusively demonstrated. By measuring mycobacterial killing activity, we evaluated IL-2-mediated functional immune enhancement ex vivo in immunological non-responders (INRs). Methods and Findings PBMC from 12 immunological non-responders (INRs) (CD4+<200/µl, HIV-RNA<50 cp/ml) on combination antiretroviral treatment (cART) were collected at baseline, and after 3 IL-2 cycles. Eight INRs receiving only cART were studied as controls. After 21 days of PBMC incubation with a virulent M. avium suspension, counts of residual colony forming units (CFUs) and concentrations of TNF-α, IL-10 and IFN-γ were determined. In IL-2 treated patients, a significant reduction in mean residual CFUs of PBMC cultures was observed (p<0.01). Moreover, following IL-2 treatment, significant increases in PBMC's IFNγ production (p = 0.02) and substantial reductions in IL-10 levels were observed. Conclusions IL-2 therapy restores the ability of the lympho-monocyte system in eliciting an effective response against mycobacterial infections. Our data indicate the possibility of a clinical role held by IL-2 in enhancing the immune function of subjects unable to achieve immune competence through cART alone.
Collapse
Affiliation(s)
- Francesca Sabbatini
- Division of Infectious Diseases, Department of Internal Medicine, San Gerardo Hospital, University of Milan-Bicocca, Monza, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Over the past decade, a family of host proteins known as suppressors of cytokine signaling (SOCS) have emerged as frequent targets of viral exploitation. Under physiologic circumstances, SOCS proteins negatively regulate inflammatory signaling pathways by facilitating ubiquitination and proteosomal degradation of pathway machinery. Their expression is tightly regulated to prevent excessive inflammation while maintaining protective antipathogenic responses. Numerous viruses, however, have developed mechanisms to induce robust host SOCS protein expression following infection, essentially "hijacking" SOCS function to promote virus survival. To date, SOCS proteins have been shown to inhibit protective antiviral signaling pathways, allowing viruses to evade the host immune response, and to ubiquitinate viral proteins, facilitating intracellular viral trafficking and progeny virus assembly. Importantly, manipulation of SOCS proteins not only facilitates progression of the viral life cycle but also powerfully shapes the presentation of viral disease. SOCS proteins can define host susceptibility to infection, contribute to peripheral disease manifestations such as immune dysfunction and cancer, and even modify the efficacy of therapeutic interventions. Looking toward the future, it is clear that a better understanding of the role of SOCS proteins in viral diseases will be essential in our struggle to modulate and even eliminate the pathogenic effects of viruses on the host.
Collapse
|
36
|
Abstract
Over the past decade, a family of host proteins known as suppressors of cytokine signaling (SOCS) have emerged as frequent targets of viral exploitation. Under physiologic circumstances, SOCS proteins negatively regulate inflammatory signaling pathways by facilitating ubiquitination and proteosomal degradation of pathway machinery. Their expression is tightly regulated to prevent excessive inflammation while maintaining protective antipathogenic responses. Numerous viruses, however, have developed mechanisms to induce robust host SOCS protein expression following infection, essentially "hijacking" SOCS function to promote virus survival. To date, SOCS proteins have been shown to inhibit protective antiviral signaling pathways, allowing viruses to evade the host immune response, and to ubiquitinate viral proteins, facilitating intracellular viral trafficking and progeny virus assembly. Importantly, manipulation of SOCS proteins not only facilitates progression of the viral life cycle but also powerfully shapes the presentation of viral disease. SOCS proteins can define host susceptibility to infection, contribute to peripheral disease manifestations such as immune dysfunction and cancer, and even modify the efficacy of therapeutic interventions. Looking toward the future, it is clear that a better understanding of the role of SOCS proteins in viral diseases will be essential in our struggle to modulate and even eliminate the pathogenic effects of viruses on the host.
Collapse
|
37
|
Akhtar LN, Qin H, Muldowney MT, Yanagisawa LL, Kutsch O, Clements JE, Benveniste EN. Suppressor of cytokine signaling 3 inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. THE JOURNAL OF IMMUNOLOGY 2010; 185:2393-404. [PMID: 20631305 DOI: 10.4049/jimmunol.0903563] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HIV-1 replication within macrophages of the CNS often results in cognitive and motor impairment, which is known as HIV-associated dementia (HAD) in its most severe form. IFN-beta suppresses viral replication within these cells during early CNS infection, but the effect is transient. HIV-1 eventually overcomes this protective innate immune response to resume replication through an unknown mechanism, initiating the progression toward HAD. In this article, we show that Suppressor of Cytokine Signaling (SOCS)3, a molecular inhibitor of IFN signaling, may allow HIV-1 to evade innate immunity within the CNS. We found that SOCS3 is elevated in an in vivo SIV/macaque model of HAD and that the pattern of expression correlates with recurrence of viral replication and onset of CNS disease. In vitro, the HIV-1 regulatory protein transactivator of transcription induces SOCS3 in human and murine macrophages in a NF-kappaB-dependent manner. SOCS3 expression attenuates the response of macrophages to IFN-beta at proximal levels of pathway activation and downstream antiviral gene expression and consequently overcomes the inhibitory effect of IFN-beta on HIV-1 replication. These studies indicate that SOCS3 expression, induced by stimuli present in the HIV-1-infected brain, such as transactivator of transcription, inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. This consequence of SOCS3 expression in vitro, supported by a correlation with increased viral load and onset of CNS disease in vivo, suggests that SOCS3 may allow HIV-1 to evade the protective innate immune response within the CNS, allowing the recurrence of viral replication and, ultimately, promoting progression toward HAD.
Collapse
|
38
|
Drevets DA, Schawang JE, Mandava VK, Dillon MJ, Leenen PJM. Severe Listeria monocytogenes infection induces development of monocytes with distinct phenotypic and functional features. THE JOURNAL OF IMMUNOLOGY 2010; 185:2432-41. [PMID: 20631315 DOI: 10.4049/jimmunol.1000486] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Monocytes perform diverse roles during infection with the facultative intracellular bacterium Listeria monocytogenes. They are essential as bactericidal cells in host defense but can also become Trojan horses transporting bacteria into the brain. To explain these contrasting roles, we characterized bone marrow (BM) monocytes in steady state and generated during lethal and sublethal L. monocytogenes infection. Ly-6C(high)CD11b(+) BM monocytes expressed high amounts of M-CSFR/CD115 in steady state and 72 h following sublethal infection. However, infection with increasing numbers of bacteria resulted in progressive loss of CD115 and strongly decreased CD115-encoding c-fms mRNA expression. Conversely, analysis of regulatory molecules showed de novo expression of the nonsignaling IL-1RII, CD121b, under the same conditions. Ly-6C(high)CD11b(+) monocytes in circulation also acquired a CD115(neg/low)CD121b(high) phenotype during lethal infection. These BM monocytes showed upregulation of suppressor of cytokine signaling 1 and 3 and IL-1R-"associated kinase-M to a greater extent and/or earlier compared with cells from sublethal infection and showed decreased LPS-induced IL-6 production despite similar levels of surface TLR4 expression. BM monocytes from uninfected or sublethally infected mice bound and internalized very few L. monocytogenes in vitro. However, both functions were significantly increased in monocytes developing during lethal infection. Nonetheless, these cells did not produce reactive oxygen intermediates, suggesting an inability to kill L. monocytogenes. Together, these data show that systemic infections with lethal and sublethal amounts of bacteria differentially shape developing BM monocytes. This results in distinct phenotypic and functional properties consistent with being Trojan horses rather than bactericidal effector cells.
Collapse
Affiliation(s)
- Douglas A Drevets
- Department of Medicine, Oklahoma University Health Sciences Center, Oklahoma City, OK 73014, USA.
| | | | | | | | | |
Collapse
|
39
|
Harding CV, Boom WH. Regulation of antigen presentation by Mycobacterium tuberculosis: a role for Toll-like receptors. Nat Rev Microbiol 2010; 8:296-307. [PMID: 20234378 PMCID: PMC3037727 DOI: 10.1038/nrmicro2321] [Citation(s) in RCA: 313] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mycobacterium tuberculosis survives in antigen-presenting cells (APCs) such as macrophages and dendritic cells. APCs present antigens in association with major histocompatibility complex (MHC) class II molecules to stimulate CD4(+) T cells, and this process is essential to contain M. tuberculosis infection. Immune evasion allows M. tuberculosis to establish persistent or latent infection in macrophages and results in Toll-like receptor 2 (TLR2)-dependent inhibition of MHC class II transactivator expression, MHC class II molecule expression and antigen presentation. This reduction of antigen presentation might reflect a general mechanism of negative-feedback regulation that prevents excessive T cell-mediated inflammation and that M. tuberculosis has subverted to create a niche for survival in infected macrophages and evasion of recognition by CD4(+) T cells.
Collapse
Affiliation(s)
- Clifford V Harding
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106-7288, USA.
| | | |
Collapse
|
40
|
Impairment of gamma interferon signaling in human neutrophils infected with Anaplasma phagocytophilum. Infect Immun 2009; 78:358-63. [PMID: 19858302 DOI: 10.1128/iai.01005-09] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Anaplasma phagocytophilum, the causative agent of tick-borne human granulocytic anaplasmosis (HGA), is an intracellular bacterium which survives and multiplies inside polymorphonuclear neutrophil granulocytes (PMN). Increased bacterial burden in gamma interferon (IFN-gamma)-deficient mice suggested a major role of IFN-gamma in the control of A. phagocytophilum. Here we investigated whether infection of human PMN with A. phagocytophilum impairs IFN-gamma signaling thus facilitating intracellular survival of the bacterium. The secretion of the IFN-gamma-inducible chemokines IP-10/CXCL10 and MIG/CXCL9 was markedly inhibited in infected neutrophils. Molecular analyses revealed that, compared to uninfected PMN, A. phagocytophilum decreased the expression of the IFN-gamma receptor alpha-chain CD119, diminished the IFN-gamma-induced phosphorylation of STAT1, and enhanced the expression of SOCS1 and SOCS3 in PMN. Since IFN-gamma activates various antibacterial effector mechanisms of PMN, the impaired IFN-gamma signaling in infected cells likely contributes to the survival of A. phagocytophilum inside PMN and to HGA disease development.
Collapse
|
41
|
Narayana Y, Bansal K, Sinha AY, Kapoor N, Puzo G, Gilleron M, Balaji KN. SOCS3 expression induced by PIM2 requires PKC and PI3K signaling. Mol Immunol 2009; 46:2947-54. [DOI: 10.1016/j.molimm.2009.06.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 06/11/2009] [Accepted: 06/18/2009] [Indexed: 10/20/2022]
|
42
|
Palma C, Iona E, Ebensen T, Guzman CA, Cassone A. The toll-like receptor 2/6 ligand MALP-2 reduces the viability of Mycobacterium tuberculosis in murine macrophages. Open Microbiol J 2009; 3:47-52. [PMID: 19471610 PMCID: PMC2685717 DOI: 10.2174/1874285800903010047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 03/17/2009] [Accepted: 03/18/2009] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptors (TLRs) sense conserved structures of pathogens and influence macrophage functions. Here we investigated the impact of TLR signaling on the modulation of macrophage defense mechanisms against infection of Mycobacterium tuberculosis (MTB), the causative agent of tuberculosis. We found that a synthetic derivative of the TLR2/6 agonist MALP-2 and the potent TLR4 agonist lipopolysaccharide inhibited the intracellular growth of MTB in murine macrophages. Likely the microbicidal effect was mediated by production of nitric oxide while it is still unclear the role played by release of TNF-α , IL-6, MIP-1β and IL-10. These results suggest that the activation of microbicidal defense via TLR ligands is an appealing target for the establishment on immune intervention against tuberculosis.
Collapse
Affiliation(s)
- Carla Palma
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | |
Collapse
|
43
|
Gratz N, Siller M, Schaljo B, Pirzada ZA, Gattermeier I, Vojtek I, Kirschning CJ, Wagner H, Akira S, Charpentier E, Kovarik P. Group A streptococcus activates type I interferon production and MyD88-dependent signaling without involvement of TLR2, TLR4, and TLR9. J Biol Chem 2008; 283:19879-87. [PMID: 18480050 PMCID: PMC2459277 DOI: 10.1074/jbc.m802848200] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Indexed: 01/07/2023] Open
Abstract
Bacterial pathogens are recognized by the innate immune system through pattern recognition receptors, such as Toll-like receptors (TLRs). Engagement of TLRs triggers signaling cascades that launch innate immune responses. Activation of MAPKs and NF-kappaB, elements of the major signaling pathways induced by TLRs, depends in most cases on the adaptor molecule MyD88. In addition, Gram-negative or intracellular bacteria elicit MyD88-independent signaling that results in production of type I interferon (IFN). Here we show that in mouse macrophages, the activation of MyD88-dependent signaling by the extracellular Gram-positive human pathogen group A streptococcus (GAS; Streptococcus pyogenes) does not require TLR2, a receptor implicated in sensing of Gram-positive bacteria, or TLR4 and TLR9. Redundant engagement of either of these TLR molecules was excluded by using TLR2/4/9 triple-deficient macrophages. We further demonstrate that infection of macrophages by GAS causes IRF3 (interferon-regulatory factor 3)-dependent, MyD88-independent production of IFN. Surprisingly, IFN is induced also by GAS lacking slo and sagA, the genes encoding cytolysins that were shown to be required for IFN production in response to other Gram-positive bacteria. Our data indicate that (i) GAS is recognized by a MyD88-dependent receptor other than any of those typically used by bacteria, and (ii) GAS as well as GAS mutants lacking cytolysin genes induce type I IFN production by similar mechanisms as bacteria requiring cytoplasmic escape and the function of cytolysins.
Collapse
Affiliation(s)
- Nina Gratz
- Max F Perutz Laboratories, Department of Microbiology and Immunobiology, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Yang T, Stark P, Janik K, Wigzell H, Rottenberg ME. SOCS-1 protects against Chlamydia pneumoniae-induced lethal inflammation but hampers effective bacterial clearance. THE JOURNAL OF IMMUNOLOGY 2008; 180:4040-9. [PMID: 18322213 DOI: 10.4049/jimmunol.180.6.4040] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Suppressor of cytokine signaling 1 (SOCS1) plays a major role in the inhibition of STAT1-mediated responses. STAT1-dependent responses are critical for resistance against infection with Chlamydia pneumoniae. We studied the regulation of expression of SOCS1 and SOCS3, and the role of SOCS1 during infection with C. pneumoniae in mice. Bone marrow-derived macrophages (BMM) and dendritic cells in vitro or lungs in vivo all showed enhanced STAT1-dependent SOCS1 mRNA accumulation after infection with C. pneumoniae. Infection-increased SOCS1 mRNA levels were dependent on IFN-alphabeta but not on IFN-gamma. T or B cells were not required for SOCS1 mRNA accumulation in vivo. Infection-induced STAT1-phosphorylation occurred more rapidly in SOCS1(-/-) BMM. In agreement, expression of IFN-gamma responsive genes, but not IL-1beta, IL-6, or TNF-alpha were relatively increased in C. pneumoniae-infected SOCS1(-/-) BMM. Surprisingly, C. pneumoniae infection-induced IFN-alpha, IFN-beta, and IFN-gamma expression in BMM were attenuated by SOCS1. C. pneumoniae infection of RAG1(-/-)/SOCS1(-/-) mice induced a rapid lethal inflammation, accompanied by diminished pulmonary bacterial load and increased levels of iNOS and IDO but not IL-1beta, IL-6, or TNF-alpha mRNA. In summary, C. pneumoniae infection induces a STAT1, IFN-alphabeta-dependent and IFN-gamma independent SOCS1 mRNA accumulation. Presence of SOCS1 controls the infection-induced lethal inflammatory disease but impairs the bacterial control.
Collapse
Affiliation(s)
- Tangbin Yang
- Department of Microbiology, Tumorbiology and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
45
|
Mycobacterial interaction with innate receptors: TLRs, C-type lectins, and NLRs. Curr Opin Infect Dis 2008; 21:279-86. [DOI: 10.1097/qco.0b013e3282f88b5d] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Narayana Y, Balaji KN. NOTCH1 up-regulation and signaling involved in Mycobacterium bovis BCG-induced SOCS3 expression in macrophages. J Biol Chem 2008; 283:12501-11. [PMID: 18332140 DOI: 10.1074/jbc.m709960200] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Suppressor of cytokine signaling (SOCS) 3 is a critical negative regulator of cytokine signaling and is induced by Mycobacterium bovis Bacille Calmette-Guérin (M. bovis BCG) in mouse macrophages. However, little is known about the early receptor proximal signaling mechanisms underlying mycobacteria-mediated induction of SOCS3. We demonstrate here for the first time that M. bovis BCG up-regulates NOTCH1 and activates the NOTCH1 signaling pathway, leading to the expression of SOCS3. We show that perturbing Notch signaling in infected macrophages results in the marked reduction in the expression of SOCS3. Furthermore, enforced expression of the Notch1 intracellular domain in RAW 264.7 macrophages induces the expression of SOCS3, which can be further potentiated by M. bovis BCG. The perturbation of Toll-like receptor (TLR) 2 signaling resulted in marked reduction in SOCS3 levels and expression of the NOTCH1 target gene, Hes1. The down-regulation of MyD88 resulted in a significant decrease in SOCS3 expression, implicating the role of the TLR2-MyD88 axis in M. bovis BCG-triggered signaling. However, the SOCS3 inducing ability of M. bovis BCG remains unaltered also upon infection of macrophages from TLR4-defective C3H/HeJ mice. More importantly, signaling perturbation data suggest the involvement of cross-talk among members of the phosphoinositide 3-kinase and mitogen-activated protein kinase cascades with NOTCH1 signaling in SOCS3 expression. Furthermore, SOCS3 expression requires the NOTCH1-mediated recruitment of Suppressor of Hairless (CSL) and nuclear factor-kappaB to the Socs3 promoter. Overall, these results implicate NOTCH1 signaling during inducible expression of SOCS3 following infection of macrophages with an intracellular bacillus-like M. bovis BCG.
Collapse
Affiliation(s)
- Yeddula Narayana
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
47
|
Qin H, Roberts KL, Niyongere SA, Cong Y, Elson CO, Benveniste EN. Molecular Mechanism of Lipopolysaccharide-Induced SOCS-3 Gene Expression in Macrophages and Microglia. THE JOURNAL OF IMMUNOLOGY 2007; 179:5966-76. [DOI: 10.4049/jimmunol.179.9.5966] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
48
|
Souza CD, Evanson OA, Weiss DJ. Role of the mitogen-activated protein kinase pathway in the differential response of bovine monocytes to Mycobacterium avium subsp. paratuberculosis and Mycobacterium avium subsp. avium. Microbes Infect 2007; 9:1545-52. [PMID: 18035573 DOI: 10.1016/j.micinf.2007.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 08/24/2007] [Accepted: 08/27/2007] [Indexed: 11/16/2022]
Abstract
We compared the kinetics of activation and antimicrobial activities of MAPK-p38 and MAPK-ERK in bovine monocytes infected with Mycobacterium avium subsp. paratuberculosis (MAP) and Mycobacterium avium subsp. avium (Maa). Monocytes were incubated with MAP or Maa organisms with or without a specific inhibitor of the MAPK-p38 pathway (SB203580), and MAPK phosphorylation and antimicrobial functions of monocytes were evaluated. At early time points MAPK-p38 phosphorylation was greater in MAP-infected bovine monocytes than in Maa-infected monocytes. At later time points MAPK-p38 phosphorylation by both organisms was similar. MAPKp38 phosphorylation in MAP-infected monocytes was similar to negative control cells, whereas in Maa-infected this activation remained greater than negative control cells. Increase phosphorylation MAPK-ERK was similar at all time points for both organisms. Bovine monocytes had minimal capacity to kill MAP organisms, to acidify MAP-containing phagosomes, or to form phagolysosome. Alternatively, bovine monocytes were able to kill Maa organisms. Addition of SB203580 to monocyte cultures increased phagosome acidification, phagolysosome formation, and killing of MAP and Maa organisms. Taken together these data indicate that early transient activation of MAPK-p38 in bovine mononuclear phagocytes by MAP organisms may be a key mechanism involved in the capacity of MAP to survive in bovine monocytes.
Collapse
Affiliation(s)
- Cleverson D Souza
- Department of Veterinary Science, University of Minnesota, St. Paul, MN 55108, USA
| | | | | |
Collapse
|
49
|
Humann J, Bjordahl R, Andreasen K, Lenz LL. Expression of the p60 autolysin enhances NK cell activation and is required for listeria monocytogenes expansion in IFN-gamma-responsive mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:2407-14. [PMID: 17277147 DOI: 10.4049/jimmunol.178.4.2407] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Both peptidoglycan and muropeptides potently modulate inflammatory and innate immune responses. The secreted Listeria monocytogenes p60 autolysin digests peptidoglycan and promotes bacterial infection in vivo. Here, we report that p60 contributes to bacterial subversion of NK cell activation and innate IFN-gamma production. L. monocytogenes deficient for p60 (Deltap60) competed well for expansion in mice doubly deficient for IFNAR1 and IFN-gammaR1 or singly deficient for IFN-gammaR1, but not in wild-type, IFNAR1(-/-), or TLR2(-/-) mice. The restored competitiveness of p60-deficient bacteria suggested a specific role for p60 in bacterial subversion of IFN-gamma-mediated immune responses, since in vivo expansion of three other mutant L. monocytogenes strains (DeltaActA, DeltaNamA, and DeltaPlcB) was not complemented in IFN-gammaR1(-/-) mice. Bacterial expression of p60 was not required to induce socs1, socs3, and il10 expression in infected mouse bone marrow macrophages but did correlate with enhanced production of IL-6, IL-12p70, and most strikingly IFN-gamma. The primary source of p60-dependent innate IFN-gamma was NK cells, whereas bacterial p60 expression did not significantly alter innate IFN-gamma production by T cells. The mechanism for p60-dependent NK cell stimulation was also indirect, given that treatment with purified p60 protein failed to directly activate NK cells for IFN-gamma production. These data suggest that p60 may act on infected cells to indirectly enhance NK cell activation and increase innate IFN-gamma production, which presumably promotes early bacterial expansion through its immunoregulatory effects on bystander cells. Thus, the simultaneous induction of IFN-gamma production and factors that inhibit IFN-gamma signaling may be a common strategy for misdirection of early antibacterial immunity.
Collapse
Affiliation(s)
- Jessica Humann
- Integrated Department of Immunology, National Jewish Medical and Research Center, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
50
|
Montaner LJ, Crowe SM, Aquaro S, Perno CF, Stevenson M, Collman RG. Advances in macrophage and dendritic cell biology in HIV-1 infection stress key understudied areas in infection, pathogenesis, and analysis of viral reservoirs. J Leukoc Biol 2006; 80:961-4. [PMID: 16935944 DOI: 10.1189/jlb.0806488] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The continued quest to intervene in HIV-1 infection by halting transmission, suppressing replication, or eradicating disease in infected subjects stresses the significance of dendritic cell and macrophage biology as early and persistent players in the relationship between infection and disease. As highlighted by new data and presentations at the Sixth International Workshop on HIV and Cells of Macrophage/Dendritic Lineage and Other Reservoirs, a greater emphasis is currently underway in studying the potential of targeting these cell types by intervention early in infection, better defining viral phenotypes and entry mechanisms with a more precise nomenclature system, identifying new, intrinsic cellular factors that may restrict infection within these cell types, and pursuing novel roles for macrophage activation and trafficking. Other key areas include examination of these cells as sources of viral persistence in patients, their roles in coinfection, and their metabolic function in HIV pathogenesis and drug toxicity. This issue of JLB contains reviews and original research reports from the workshop, which highlight new findings, current research questions, and key areas in need of future investigation as a result of their significance to HIV prevention and pathogenesis.
Collapse
|