1
|
Merritt EF, Kochanowsky JA, Hervé P, Watson AA, Koshy AA. Toxoplasma type II effector GRA15 has limited influence in vivo. PLoS One 2024; 19:e0300764. [PMID: 38551902 PMCID: PMC10980211 DOI: 10.1371/journal.pone.0300764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/03/2024] [Indexed: 04/01/2024] Open
Abstract
Toxoplasma gondii is an intracellular parasite that establishes a long-term infection in the brain of many warm-blooded hosts, including humans and rodents. Like all obligate intracellular microbes, Toxoplasma uses many effector proteins to manipulate the host cell to ensure parasite survival. While some of these effector proteins are universal to all Toxoplasma strains, some are polymorphic between Toxoplasma strains. One such polymorphic effector is GRA15. The gra15 allele carried by type II strains activates host NF-κB signaling, leading to the release of cytokines such as IL-12, TNF, and IL-1β from immune cells infected with type II parasites. Prior work also suggested that GRA15 promotes early host control of parasites in vivo, but the effect of GRA15 on parasite persistence in the brain and the peripheral immune response has not been well defined. For this reason, we sought to address this gap by generating a new IIΔgra15 strain and comparing outcomes at 3 weeks post infection between WT and IIΔgra15 infected mice. We found that the brain parasite burden and the number of macrophages/microglia and T cells in the brain did not differ between WT and IIΔgra15 infected mice. In addition, while IIΔgra15 infected mice had a lower number and frequency of splenic M1-like macrophages and frequency of PD-1+ CTLA-4+ CD4+ T cells and NK cells compared to WT infected mice, the IFN-γ+ CD4 and CD8 T cell populations were equivalent. In summary, our results suggest that in vivo GRA15 may have a subtle effect on the peripheral immune response, but this effect is not strong enough to alter brain parasite burden or parenchymal immune cell number at 3 weeks post infection.
Collapse
Affiliation(s)
- Emily F. Merritt
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Joshua A. Kochanowsky
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Perrine Hervé
- Microbiologie Fondamentale et Pathogénicité, CNRS UMR 5234, Université de Bordeaux, Bordeaux, France
| | - Alison A. Watson
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Anita A. Koshy
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
2
|
Andualem H, Hollams E, Kollmann TR, Amenyogbe N. BCG-Induced Immune Training: Interplay between Trained Immunity and Emergency Granulopoiesis. J Mol Biol 2023; 435:168169. [PMID: 37263392 PMCID: PMC11688642 DOI: 10.1016/j.jmb.2023.168169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023]
Abstract
Bacille Calmette-Guérin (BCG) is the most commonly administered vaccine in human history. The medical application of BCG extends far beyond the fight against tuberculosis. Despite its stellar medical record over 100 years, insight into how BCG provides this vast range of benefits is largely limited, both for its pathogen-specific (tuberculosis) as well as pathogen-agnostic (other infections, autoimmunity, allergies, and cancer) effects. Trained immunity and emergency granulopoiesis have been identified as mediating BCG's pathogen-agnostic effects, for which some of the molecular mechanisms have been delineated. Upon review of the existing evidence, we postulate that emergency granulopoiesis and trained immunity are a continuum of the same effect cascade. In this context, we highlight that BCG's pathogen-agnostic benefits could be optimized by taking advantage of the age of the recipient and route of BCG administration.
Collapse
Affiliation(s)
- Henok Andualem
- Department of Medical Laboratory Science, College of Health Science, Debre Tabor University, Ethiopia.
| | - Elysia Hollams
- Telethon Kids Institute, Perth, Western Australia, Australia
| | | | - Nelly Amenyogbe
- Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
3
|
Xue T, Lu Z, Zhang W, Wang Z, Shi Y, Jiang H, Wang H. Facial Erythema Due to Lupus Vulgaris and Candida albicans Infections: A Case Report. Clin Cosmet Investig Dermatol 2022; 15:1397-1402. [PMID: 35910508 PMCID: PMC9325874 DOI: 10.2147/ccid.s372359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022]
Abstract
Co-infection of Mycobacterium tuberculosis (MTB) and Candida albicans with erythema on the face is rare. A familiar red spot on the face can easily lead to missed diagnosis and misdiagnosis. Untreated lupus vulgaris (LV) can form scar tissue. And the fungal infection that cannot be diagnosed and treated timely can also lead to failure of LV treatment, resulting in facial scarring, disfigurement, and psychological stress. In this study, we reported a case of a 58-year-old immunocompetent female co-infected with MTB and Candida albicans on her face. After anti-tuberculous and anti-fungal therapy, she recovered with no scar on her face.
Collapse
Affiliation(s)
- Tianping Xue
- Department of Dermatology, Suzhou Wuzhong People's Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Zhenzhong Lu
- Department of Dermatology, Suzhou Wuzhong People's Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Wenyue Zhang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Zhenzhen Wang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Ying Shi
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Haiqin Jiang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Hongsheng Wang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.,Centre for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
4
|
Evaluation of early innate and adaptive immune responses to the TB vaccine Mycobacterium bovis BCG and vaccine candidate BCGΔBCG1419c. Sci Rep 2022; 12:12377. [PMID: 35858977 PMCID: PMC9300728 DOI: 10.1038/s41598-022-14935-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/03/2022] [Indexed: 12/30/2022] Open
Abstract
The vaccine Mycobacterium bovis Bacillus Calmette-Guérin (BCG) elicits an immune response that is protective against certain forms of tuberculosis (TB); however, because BCG efficacy is limited it is important to identify alternative TB vaccine candidates. Recently, the BCG deletion mutant and vaccine candidate BCGΔBCG1419c was demonstrated to survive longer in intravenously infected BALB/c mice due to enhanced biofilm formation, and better protected both BALB/c and C57BL/6 mice against TB-induced lung pathology during chronic stages of infection, relative to BCG controls. BCGΔBCG1419c-elicited protection also associated with lower levels of proinflammatory cytokines (i.e. IL6, TNFα) at the site of infection in C57BL/6 mice. Given the distinct immune profiles of BCG- and BCGΔBCG1419c-immunized mice during chronic TB, we set out to determine if there are early immunological events which distinguish these two groups, using multi-dimensional flow cytometric analysis of the lungs and other tissues soon after immunization. Our results demonstrate a number of innate and adaptive response differences between BCG- and BCGΔBCG1419c-immunized mice which are consistent with the latter being longer lasting and potentially less inflammatory, including lower frequencies of exhausted CD4+ T helper (TH) cells and higher frequencies of IL10-producing T cells, respectively. These studies suggest the use of BCGΔBCG1419c may be advantageous as an alternative TB vaccine candidate.
Collapse
|
5
|
Brook B, Schaltz-Buchholzer F, Ben-Othman R, Kollmann T, Amenyogbe N. A place for neutrophils in the beneficial pathogen-agnostic effects of the BCG vaccine. Vaccine 2022; 40:1534-1539. [PMID: 33863572 PMCID: PMC11688641 DOI: 10.1016/j.vaccine.2021.03.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/25/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022]
Abstract
The BCG vaccine has long been recognized for reducing the risk to suffer from infectious diseases unrelated to its target disease, tuberculosis. Evidence from human trials demonstrate substantial reductions in all-cause mortality, especially in the first week of life. Observational studies have identified an association between BCG vaccination and reduced risk of respiratory infectious disease and clinical malaria later in childhood. The mechanistic basis for these pathogen-agnostic benefits, also known as beneficial non-specific effects (NSE) of BCG have been attributed to trained immunity, or epigenetic reprogramming of hematopoietic cells that give rise to innate immune cells responding more efficiently to a broad range of pathogens. Furthermore, within trained immunity, the focus so far has been on enhanced monocyte function. However, polymorphonuclear cells, namely neutrophils, are not only major constituents of the hematopoietic compartment but functionally as well as numerically represent a prominent component of the immune system. The beneficial NSEs of the BCG vaccine on newborn sepsis was recently demonstrated to be driven by a BCG-mediated numeric increase of neutrophils (emergency granulopoiesis (EG)). And experimental evidence in animal models suggest that BCG can modulate neutrophil function as well. Together, these findings suggest that neutrophils are crucial to at least the immediate beneficial NSE of the BCG vaccine. Efforts to uncover the full gamut of mechanisms underpinning the broad beneficial effects of BCG should therefore include neutrophils at the forefront.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Frederick Schaltz-Buchholzer
- Institute of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark; Bandim Health Project, INDEPTH Network, Bissau, Guinea-Bissau
| | - Rym Ben-Othman
- Telethon Kids Institute, Perth, Western Australia, Australia
| | - Tobias Kollmann
- Telethon Kids Institute, Perth, Western Australia, Australia
| | - Nelly Amenyogbe
- Telethon Kids Institute, Perth, Western Australia, Australia.
| |
Collapse
|
6
|
Larsen MH, Lacourciere K, Parker TM, Kraigsley A, Achkar JM, Adams LB, Dupnik KM, Hall-Stoodley L, Hartman T, Kanipe C, Kurtz SL, Miller MA, Salvador LCM, Spencer JS, Robinson RT. The Many Hosts of Mycobacteria 8 (MHM8): A conference report. Tuberculosis (Edinb) 2020; 121:101914. [PMID: 32279870 PMCID: PMC7428850 DOI: 10.1016/j.tube.2020.101914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 12/18/2022]
Abstract
Mycobacteria are important causes of disease in human and animal hosts. Diseases caused by mycobacteria include leprosy, tuberculosis (TB), nontuberculous mycobacteria (NTM) infections and Buruli Ulcer. To better understand and treat mycobacterial disease, clinicians, veterinarians and scientists use a range of discipline-specific approaches to conduct basic and applied research, including conducting epidemiological surveys, patient studies, wildlife sampling, animal models, genetic studies and computational simulations. To foster the exchange of knowledge and collaboration across disciplines, the Many Hosts of Mycobacteria (MHM) conference series brings together clinical, veterinary and basic scientists who are dedicated to advancing mycobacterial disease research. Started in 2007, the MHM series recently held its 8th conference at the Albert Einstein College of Medicine (Bronx, NY). Here, we review the diseases discussed at MHM8 and summarize the presentations on research advances in leprosy, NTM and Buruli Ulcer, human and animal TB, mycobacterial disease comorbidities, mycobacterial genetics and 'omics, and animal models. A mouse models workshop, which was held immediately after MHM8, is also summarized. In addition to being a resource for those who were unable to attend MHM8, we anticipate this review will provide a benchmark to gauge the progress of future research concerning mycobacteria and their many hosts.
Collapse
Affiliation(s)
- Michelle H Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Karen Lacourciere
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Tina M Parker
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | - Alison Kraigsley
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Jacqueline M Achkar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Linda B Adams
- Department of Health and Human Services, Health Resources and Services Administration, Healthcare Systems Bureau, National Hansen's Disease Programs, Baton Rouge, LA, USA
| | - Kathryn M Dupnik
- Center for Global Health, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Luanne Hall-Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Travis Hartman
- Center for Global Health, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Carly Kanipe
- Department of Immunobiology, Iowa State University, Ames, IA, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA; Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Sherry L Kurtz
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Washington, DC, USA
| | - Michele A Miller
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Liliana C M Salvador
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA; Institute of Bioinformatics, University of Georgia, Athens, GA, USA; Center for the Ecology of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - John S Spencer
- Department of Microbiology, Immunology, and Pathology, Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO, USA
| | - Richard T Robinson
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
7
|
Merkhofer RM, Klein BS. Advances in Understanding Human Genetic Variations That Influence Innate Immunity to Fungi. Front Cell Infect Microbiol 2020; 10:69. [PMID: 32185141 PMCID: PMC7058545 DOI: 10.3389/fcimb.2020.00069] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/12/2020] [Indexed: 12/30/2022] Open
Abstract
Fungi are ubiquitous. Yet, despite our frequent exposure to commensal fungi of the normal mammalian microbiota and environmental fungi, serious, systemic fungal infections are rare in the general population. Few, if any, fungi are obligate pathogens that rely on infection of mammalian hosts to complete their lifecycle; however, many fungal species are able to cause disease under select conditions. The distinction between fungal saprophyte, commensal, and pathogen is artificial and heavily determined by the ability of an individual host's immune system to limit infection. Dramatic examples of commensal fungi acting as opportunistic pathogens are seen in hosts that are immune compromised due to congenital or acquired immune deficiency. Genetic variants that lead to immunological susceptibility to fungi have long been sought and recognized. Decreased myeloperoxidase activity in neutrophils was first reported as a mechanism for susceptibility to Candida infection in 1969. The ability to detect genetic variants and mutations that lead to rare or subtle susceptibilities has improved with techniques such as single nucleotide polymorphism (SNP) microarrays, whole exome sequencing (WES), and whole genome sequencing (WGS). Still, these approaches have been limited by logistical considerations and cost, and they have been applied primarily to Mendelian impairments in anti-fungal responses. For example, loss-of-function mutations in CARD9 were discovered by studying an extended family with a history of fungal infection. While discovery of such mutations furthers the understanding of human antifungal immunity, major Mendelian susceptibility loci are unlikely to explain genetic disparities in the rate or severity of fungal infection on the population level. Recent work using unbiased techniques has revealed, for example, polygenic mechanisms contributing to candidiasis. Understanding the genetic underpinnings of susceptibility to fungal infections will be a powerful tool in the age of personalized medicine. Future application of this knowledge may enable targeted health interventions for susceptible individuals, and guide clinical decision making based on a patient's individual susceptibility profile.
Collapse
Affiliation(s)
- Richard M Merkhofer
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Bruce S Klein
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Department of Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
8
|
Wu CY, Chiu HY, Tsai TF. The seroconversion rate of QuantiFERON-TB Gold In-Tube test in psoriatic patients receiving secukinumab and ixekizumab, the anti-interleukin-17A monoclonal antibodies. PLoS One 2019; 14:e0225112. [PMID: 31881026 PMCID: PMC6934285 DOI: 10.1371/journal.pone.0225112] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND For psoriatic patients receiving biologics, the concern of tuberculosis (TB) infection exists. Although the TB risk of anti-interleukin (IL)-17A agents is generally considered very low, more real-world data are needed to support the safety. OBJECTIVES This study aims to provide the real-world experience of using serial QuantiFERON-TB Gold In-Tube (QFT-GIT) test among patients treated with secukinumab or ixekizumab in Taiwan, an intermediate TB burden country, for the detection of latent TB infection (LTBI) reactivation or newly acquired TB infection. METHODS This retrospective review evaluated 100 consecutive patients with psoriasis receiving anti-IL-17A therapies who were checked with at least twice QFT-GIT between 2016 and 2019 in National Taiwan University Hospital, Taipei and Hsin-Chu, Taiwan. RESULTS Among the 100 patients, the baseline QFT-GIT results were negative in 81.0% (81/100), positive in 18.0% (18/100), and indeterminate in 1.0% (1/100) of patients. The overall outcomes in patients receiving at least 6 months of cumulative exposure to anti-IL-17A agents were persistently seronegative in 80 patients (80.0%), persistently seropositive in 14 patients (14.0%), seroconversion in 1 patient (1.0%), seroreversion in 3 patients (3.0%), and others in 2 patients (2.0%). In patients with at least 11 months of cumulative exposure, the seroconversion rate was 1.3% (1/79). The only case with seroconversion had a positive QFT-GIT result previously. No case of TB reactivation or newly acquired TB infection was identified during the follow-up. CONCLUSIONS In patients treated with anti-IL-17A monoclonal antibodies for psoriasis, routine serial repeat QFT-GIT testing was associated with lower seroconversion rate compared to real-world data of tumor necrosis factor-α inhibitors and anti-IL-12/23 antibody in Taiwan and in pivotal studies. Because clinical TB symptoms and signs are often preceded by QFT-GIF seroconversion, this result further supports the safety of anti-IL-17A agents in patients with psoriasis for LTBI.
Collapse
Affiliation(s)
- Chen-Yu Wu
- Department of Dermatology, Cathay General Hospital, Taipei, Taiwan
| | - Hsien-Yi Chiu
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Dermatology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
9
|
Ribero S, Licciardello M, Quaglino P, Dapavo P. Efficacy and Safety of Secukinumab in Patients with Plaque Psoriasis and Latent Tuberculosis. Case Rep Dermatol 2019; 11:23-28. [PMID: 31662735 PMCID: PMC6816124 DOI: 10.1159/000501989] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/30/2022] Open
Abstract
Upon the association of biologic treatments with reactivation of latent tuberculosis infection (LTBI), screening for Mycobacterium tuberculosisinfection and anti-tuberculosis chemoprophylaxis in positive patients are required prior to biologic drug administration. Nevertheless, the risk of infection relapses associated with biologic drugs seems to be different. No cases of reactivation of LTBI have been observed in secukinumab-treated subjects, in contrast with clinical reports on the risk associated with anti-tumor necrosis factor Α-based therapy. Twelve patients with moderate to severe plaque psoriasis eligible for systemic treatment and found to have LTBI received secukinumab without previous chemoprophylaxis initiation because of clinical contraindication for 10 cases and refusal by 2 patients. None of them had tuberculosis reactivation.
Collapse
Affiliation(s)
- Simone Ribero
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| | - Matteo Licciardello
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| | - Pietro Quaglino
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| | - Paolo Dapavo
- Medical Sciences Department, Section of Dermatology, University of Turin, Turin, Italy
| |
Collapse
|
10
|
Candida albicans Elicits Pro-Inflammatory Differential Gene Expression in Intestinal Peyer's Patches. Mycopathologia 2019; 184:461-478. [PMID: 31230200 DOI: 10.1007/s11046-019-00349-4] [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: 02/01/2019] [Accepted: 06/05/2019] [Indexed: 12/17/2022]
Abstract
The details of how gut-associated lymphoid tissues such as Peyer's patches (PPs) in the small intestine play a role in immune surveillance, microbial differentiation and the mucosal barrier protection in response to fungal organisms such as Candida albicans are still unclear. We particularly focus on PPs as they are the immune sensors and inductive sites of the gut that influence inflammation and tolerance. We have previously demonstrated that CD11c+ phagocytes that include dendritic cells and macrophages are located in the sub-epithelial dome within PPs sample C. albicans. To gain insight on how specific cells within PPs sense and respond to the sampling of fungi, we gavaged naïve mice with C. albicans strains ATCC 18804 and SC5314 as well as Saccharomyces cerevisiae. We measured the differential gene expression of sorted CD45+ B220+ B-cells, CD3+ T-cells and CD11c+ DCs within the first 24 h post-gavage using nanostring nCounter® technology. The results reveal that at 24 h, PP phagocytes were the cell type that displayed differential gene expression. These phagocytes were able to sample C. albicans and discriminate between strains. In particular, strain ATCC 18804 upregulated fungal-specific pro-inflammatory genes pertaining to innate and adaptive immune responses. Interestingly, PP CD11c+ phagocytes also differentially expressed genes in response to C. albicans that were important in the protection of the mucosal barrier. These results highlight that the mucosal barrier not only responds to C. albicans, but also aids in the protection of the host.
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Many genetic conditions predispose affected individuals to opportunistic infections. A number of immunodeficiency diseases, including genetic defects termed Mendelian susceptibility to mycobacterial disease (MSMD), permit infection from many different strains of mycobacteria that would otherwise not cause disease. These include tuberculous and nontuberculous mycobacteria, and bacille Calmette-Guérin vaccine (BCG). Patients may present with infections from other organisms that depend on macrophage function for containment. Defects in multiple genes in the IL-12 and NFKB signaling pathways can cause the MSMD phenotype, some of which include IL12RB1, IL12B, IKBKG, ISG15, IFNGR1, IFNGR2, CYBB, TYK2, IRF8, and STAT1. RECENT FINDINGS Multiple autosomal recessive and dominant, and 2 X-linked recessive gene defects resulting in the MSMD phenotype have been reported, and others await discovery. This review presents the known gene defects and describes clinical findings that result from the mutations. If MSMD is suspected, a careful clinical history and examination and basic immunodeficiency screening tests will narrow the differential diagnosis. A specific diagnosis requires more sophisticated laboratory investigation. Genetic testing permits a definitive diagnosis, permitting genetic counseling. Mild cases respond well to appropriate antibiotic therapy, whereas severe disease may require hematopoietic stem cell transplantation.
Collapse
|
12
|
Abstract
Nontuberculous mycobacteria (NTM) include species that colonize human epithelia, as well as species that are ubiquitous in soil and aquatic environments. NTM that primarily inhabit soil and aquatic environments include the Mycobacterium avium complex (MAC, M. avium and Mycobacterium intracellulare) and the Mycobacterium abscessus complex (MABSC, M. abscessus subspecies abscessus, massiliense, and bolletii), and can be free-living, biofilm-associated, or amoeba-associated. Although NTM are rarely pathogenic in immunocompetent individuals, those who are immunocompromised - due to either an inherited or acquired immunodeficiency - are highly susceptible to NTM infection (NTMI). Several characteristics such as biofilm formation and the ability of select NTM species to form distinct colony morphotypes all may play a role in pathogenesis not observed in the related, well-characterized pathogen Mycobacterium tuberculosis The recognition of different morphotypes of NTM has been established and characterized since the 1950s, but the mechanisms that underlie colony phenotype change and subsequent differences in pathogenicity are just beginning to be explored. Advances in genomic analysis have led to progress in identifying genes important to the pathogenesis and persistence of MAC disease as well as illuminating genetic aspects of different colony morphotypes. Here we review recent literature regarding NTM ecology and transmission, as well as the factors which regulate colony morphotype and pathogenicity.
Collapse
Affiliation(s)
- Tiffany A Claeys
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Richard T Robinson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
13
|
Abstract
Although classically associated with myelopoiesis, granulocyte-macrophage colony-stimulating factor (GM-CSF) is increasingly recognized as being important for tuberculosis (TB) resistance. GM-CSF is expressed by nonhematopoietic and hematopoietic lineages following infection with Mycobacterium tuberculosis and is necessary to restrict M. tuberculosis growth in experimental models. Until the recent study by Rothchild et al. (mBio 8:e01514-17, 2017, https://doi.org/10.1128/mBio.01514-17), it was unknown whether GM-CSF-producing T cells contribute to TB resistance. Rothchild et al. identify which conventional and nonconventional T cell subsets produce GM-CSF during experimental TB, establish their protective nature using a variety of approaches, and provide a mechanistic basis for their ability to restrict M. tuberculosis growth. This commentary discusses the significance of these findings to basic and applied TB research. As translated to human disease, these findings suggest vaccine-mediated expansion of GM-CSF-producing T cells could be an effective prophylactic or therapeutic TB strategy.
Collapse
|
14
|
Kammüller M, Tsai TF, Griffiths CE, Kapoor N, Kolattukudy PE, Brees D, Chibout SD, Safi J, Fox T. Inhibition of IL-17A by secukinumab shows no evidence of increased Mycobacterium tuberculosis infections. Clin Transl Immunology 2017; 6:e152. [PMID: 28868144 PMCID: PMC5579471 DOI: 10.1038/cti.2017.34] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 12/25/2022] Open
Abstract
Secukinumab, a fully human monoclonal antibody that selectively neutralizes interleukin-17A (IL-17A), has been shown to have significant efficacy in the treatment of moderate to severe psoriasis, psoriatic arthritis and ankylosing spondylitis. Blocking critical mediators of immunity may carry a risk of increased opportunistic infections. Here we present clinical and in vitro findings examining the effect of secukinumab on Mycobacterium tuberculosis infection. We re-assessed the effect of secukinumab on the incidence of acute tuberculosis (TB) and reactivation of latent TB infection (LTBI) in pooled safety data from five randomized, double-blind, placebo-controlled, phase 3 clinical trials in subjects with moderate to severe plaque psoriasis. No cases of TB were observed after 1 year. Importantly, in subjects with a history of pulmonary TB (but negative for interferon-γ release and receiving no anti-TB medication) or positive for latent TB (screened by interferon-γ release assay and receiving anti-TB medication), no cases of active TB were reported. Moreover, an in vitro study examined the effect of the anti-tumor necrosis factor-α (TNFα) antibody adalimumab and secukinumab on dormant M. tuberculosis H37Rv in a novel human three-dimensional microgranuloma model. Auramine-O, Nile red staining and rifampicin resistance of M. tuberculosis were measured. In vitro, anti-TNFα treatment showed increased staining for Auramine-O, decreased Nile red staining and decreased rifampicin resistance, indicative of mycobacterial reactivation. In contrast, secukinumab treatment was comparable to control indicating a lack of effect on M. tuberculosis dormancy. To date, clinical and preclinical investigations with secukinumab found no evidence of increased M. tuberculosis infections.
Collapse
Affiliation(s)
- Michael Kammüller
- Translational Medicine-Preclinical Safety, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Christopher Em Griffiths
- Dermatology Centre, Salford Royal Hospital, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nidhi Kapoor
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Pappachan E Kolattukudy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Dominique Brees
- Translational Medicine-Preclinical Safety, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Salah-Dine Chibout
- Translational Medicine-Preclinical Safety, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Jorge Safi
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Todd Fox
- Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|