70 results on '"Mayer-Barber KD"'
Search Results
2. Eosinophils as Modulators of Host Defense during Parasitic, Fungal, Bacterial, and Viral Infections.
- Author
-
Gazzinelli-Guimaraes PH, Jones SM, Voehringer D, Mayer-Barber KD, and Samarasinghe AE
- Abstract
Eosinophils, traditionally associated as central innate effector cells with type-2 immunity during allergic and helminth parasitic diseases, have recently been revealed to have important roles in tissue homeostasis as well as host defense in a broader variety of infectious diseases. In a dedicated session at the 2023 biennial conference of the International Eosinophil Society titled "Eosinophils in Host Defense", the multifaceted roles eosinophils play against diverse pathogens ranging from parasites to fungi, bacteria, and viruses was presented. In this review, the session speakers offer a comprehensive summary of recent discoveries across pathogen classes, positioning eosinophils as pivotal leukocytes in both host defense and pathology. By unraveling the intricacies of eosinophil engagement in host resistance, this exploration may provide valuable insights not only to understand specific underpinnings of the eosinophil functions related to each class of pathogens, but also to develop novel therapeutics effective against a broad spectrum of infectious diseases., (© The Author(s) 2024. Published by Oxford University Press on behalf of Society for Leukocyte Biology.)
- Published
- 2024
- Full Text
- View/download PDF
3. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication.
- Author
-
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, and Mayer-Barber KD
- Abstract
SARS-CoV-2 infection leads to vastly divergent clinical outcomes ranging from asymptomatic infection to fatal disease. Co-morbidities, sex, age, host genetics and vaccine status are known to affect disease severity. Yet, how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure impacts the control of viral replication remains poorly understood. We demonstrate here that immune events in the mouse lung closely preceding SARS-CoV-2 infection significantly impact viral control and we identify key innate immune pathways required to limit viral replication. A diverse set of pulmonary inflammatory stimuli, including resolved antecedent respiratory infections with S. aureus or influenza, ongoing pulmonary M. tuberculosis infection, ovalbumin/alum-induced asthma or airway administration of defined TLR ligands and recombinant cytokines, all establish an antiviral state in the lung that restricts SARS-CoV-2 replication upon infection. In addition to antiviral type I interferons, the broadly inducible inflammatory cytokines TNFα and IL-1 precondition the lung for enhanced viral control. Collectively, our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation that precedes or accompanies SARS-CoV-2 exposure may be a significant factor contributing to the population-wide variability in COVID-19 disease outcomes.
- Published
- 2024
- Full Text
- View/download PDF
4. A TNF-IL-1 circuit controls Yersinia within intestinal pyogranulomas.
- Author
-
Matsuda R, Sorobetea D, Zhang J, Peterson ST, Grayczyk JP, Yost W, Apenes N, Kovalik ME, Herrmann B, O'Neill RJ, Bohrer AC, Lanza M, Assenmacher CA, Mayer-Barber KD, Shin S, and Brodsky IE
- Subjects
- Humans, Interleukin-1, Yersinia, Tumor Necrosis Factor-alpha, Monocytes, Yersinia Infections, Yersinia pseudotuberculosis
- Abstract
Tumor necrosis factor (TNF) is a pleiotropic inflammatory cytokine that mediates antimicrobial defense and granuloma formation in response to infection by numerous pathogens. We previously reported that Yersinia pseudotuberculosis colonizes the intestinal mucosa and induces the recruitment of neutrophils and inflammatory monocytes into organized immune structures termed pyogranulomas (PG) that control Yersinia infection. Inflammatory monocytes are essential for the control and clearance of Yersinia within intestinal PG, but how monocytes mediate Yersinia restriction is poorly understood. Here, we demonstrate that TNF signaling in monocytes is required for bacterial containment following enteric Yersinia infection. We further show that monocyte-intrinsic TNFR1 signaling drives the production of monocyte-derived interleukin-1 (IL-1), which signals through IL-1 receptors on non-hematopoietic cells to enable PG-mediated control of intestinal Yersinia infection. Altogether, our work reveals a monocyte-intrinsic TNF-IL-1 collaborative inflammatory circuit that restricts intestinal Yersinia infection., (© 2024 Matsuda et al.)
- Published
- 2024
- Full Text
- View/download PDF
5. BACH1 promotes tissue necrosis and Mycobacterium tuberculosis susceptibility.
- Author
-
Amaral EP, Namasivayam S, Queiroz ATL, Fukutani E, Hilligan KL, Aberman K, Fisher L, Bomfim CCB, Kauffman K, Buchanan J, Santuo L, Gazzinelli-Guimaraes PH, Costa DL, Teixeira MA, Barreto-Duarte B, Rocha CG, Santana MF, Cordeiro-Santos M, Barber DL, Wilkinson RJ, Kramnik I, Igarashi K, Scriba T, Mayer-Barber KD, Andrade BB, and Sher A
- Subjects
- Animals, Mice, Basic-Leucine Zipper Transcription Factors genetics, Macrophages microbiology, Necrosis, Mycobacterium tuberculosis genetics, Tuberculosis microbiology, Tuberculosis, Pulmonary genetics
- Abstract
Oxidative stress triggers ferroptosis, a form of cellular necrosis characterized by iron-dependent lipid peroxidation, and has been implicated in Mycobacterium tuberculosis (Mtb) pathogenesis. We investigated whether Bach1, a transcription factor that represses multiple antioxidant genes, regulates host resistance to Mtb. We found that BACH1 expression is associated clinically with active pulmonary tuberculosis. Bach1 deletion in Mtb-infected mice increased glutathione levels and Gpx4 expression that inhibit lipid peroxidation. Bach1
-/- macrophages exhibited increased resistance to Mtb-induced cell death, while Mtb-infected Bach1-deficient mice displayed reduced bacterial loads, pulmonary necrosis and lipid peroxidation concurrent with increased survival. Single-cell RNA-seq analysis of lungs from Mtb-infected Bach1-/- mice revealed an enrichment of genes associated with ferroptosis suppression. Bach1 depletion in Mtb-infected B6.Sst1S mice that display human-like necrotic lung pathology also markedly reduced necrosis and increased host resistance. These findings identify Bach1 as a key regulator of cellular and tissue necrosis and host resistance in Mtb infection., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2024
- Full Text
- View/download PDF
6. Bacterial-induced or passively administered interferon gamma conditions the lung for early control of SARS-CoV-2.
- Author
-
Hilligan KL, Namasivayam S, Clancy CS, Baker PJ, Old SI, Peluf V, Amaral EP, Oland SD, O'Mard D, Laux J, Cohen M, Garza NL, Lafont BAP, Johnson RF, Feng CG, Jankovic D, Lamiable O, Mayer-Barber KD, and Sher A
- Subjects
- Animals, Mice, SARS-CoV-2, Interferon-gamma, Lung, COVID-19 prevention & control, Interferon Type I pharmacology
- Abstract
Type-1 and type-3 interferons (IFNs) are important for control of viral replication; however, less is known about the role of Type-2 IFN (IFNγ) in anti-viral immunity. We previously observed that lung infection with Mycobacterium bovis BCG achieved though intravenous (iv) administration provides strong protection against SARS-CoV-2 in mice yet drives low levels of type-1 IFNs but robust IFNγ. Here we examine the role of ongoing IFNγ responses to pre-established bacterial infection on SARS-CoV-2 disease outcomes in two murine models. We report that IFNγ is required for iv BCG induced reduction in pulmonary viral loads, an outcome dependent on IFNγ receptor expression by non-hematopoietic cells. Importantly, we show that BCG infection prompts pulmonary epithelial cells to upregulate IFN-stimulated genes with reported anti-viral activity in an IFNγ-dependent manner, suggesting a possible mechanism for the observed protection. Finally, we confirm the anti-viral properties of IFNγ by demonstrating that the recombinant cytokine itself provides strong protection against SARS-CoV-2 challenge when administered intranasally. Together, our data show that a pre-established IFNγ response within the lung is protective against SARS-CoV-2 infection, suggesting that concurrent or recent infections that drive IFNγ may limit the pathogenesis of SARS-CoV-2 and supporting possible prophylactic uses of IFNγ in COVID-19 management., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply 2023.)
- Published
- 2023
- Full Text
- View/download PDF
7. Endogenous and Therapeutic 25-Hydroxycholesterols May Worsen Early SARS-CoV-2 Pathogenesis in Mice.
- Author
-
Fessler MB, Madenspacher JH, Baker PJ, Hilligan KL, Bohrer AC, Castro E, Meacham J, Chen SH, Johnson RF, McDonald JG, Martin NP, Tucker CJ, Mahapatra D, Cesta M, and Mayer-Barber KD
- Subjects
- Humans, Animals, Mice, SARS-CoV-2, Herpesvirus 4, Human, Hydroxycholesterols pharmacology, Cholesterol, Receptors, G-Protein-Coupled, Antiviral Agents pharmacology, Cytokines, Weight Loss, Epstein-Barr Virus Infections, COVID-19
- Abstract
Oxysterols (i.e., oxidized cholesterol species) have complex roles in biology. 25-Hydroxycholesterol (25HC), a product of the activity of cholesterol-25-hydroxylase (CH25H) on cholesterol, has recently been shown to be broadly antiviral, suggesting therapeutic potential against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, 25HC can also amplify inflammation and be converted by CYP7B1 (cytochrome P450 family 7 subfamily B member 1) to 7α,25-dihydroxycholesterol, a lipid with chemoattractant activity, via the G protein-coupled receptor EBI2 (Epstein-Barr virus-induced gene 2)/GPR183 (G protein-coupled receptor 183). Here, using in vitro studies and two different murine models of SARS-CoV-2 infection, we investigate the effects of these two oxysterols on SARS-CoV-2 pneumonia. We show that although 25HC and enantiomeric-25HC are antiviral in vitro against human endemic coronavirus-229E, they did not inhibit SARS-CoV-2; nor did supplemental 25HC reduce pulmonary SARS-CoV-2 titers in the K18-human ACE2 (angiotensin-converting enzyme 2) mouse model in vivo . Treatment with 25HC also did not alter immune cell influx into the airway, airspace cytokines, lung pathology, weight loss, symptoms, or survival but was associated with increased airspace albumin, an indicator of microvascular injury, and increased plasma proinflammatory cytokines. Conversely, mice treated with the EBI2/GPR183 inhibitor NIBR189 displayed a modest increase in lung viral load only at late time points but no change in weight loss. Consistent with these findings, although Ch25h and 25HC were upregulated in the lungs of SARS-CoV-2-infected wild-type mice, lung viral titers and weight loss in Ch25h
-/- and Gpr183-/- mice infected with the β variant were similar to those in control animals. Taken together, endogenous 25HCs do not significantly regulate early SARS-CoV-2 replication or pathogenesis, and supplemental 25HC may have proinjury rather than therapeutic effects in SARS-CoV-2 pneumonia.- Published
- 2023
- Full Text
- View/download PDF
8. IRGM1 supports host defense against intracellular bacteria through suppression of type I interferon in mice.
- Author
-
Rai P, Sharpe M, Ganta CK, Baker PJ, Mayer-Barber KD, Fee BE, Taylor GA, and Fessler MB
- Subjects
- Animals, Mice, Bacteria, Immunity, Innate, Macrophages microbiology, Interferon Type I
- Published
- 2023
- Full Text
- View/download PDF
9. Granulocytes subsets and their divergent functions in host resistance to Mycobacterium tuberculosis - a 'tipping-point' model of disease exacerbation.
- Author
-
Mayer-Barber KD
- Subjects
- Humans, Neutrophils, Eosinophils, Immunity, Innate, Mycobacterium tuberculosis, Tuberculosis
- Abstract
Granulocytes are innate immune effector cells with essential functions in host resistance to bacterial infections. I will discuss emerging evidence that during Mycobacterium tuberculosis infection, counter-intuitively, eosinophils are host-protective while neutrophils are host detrimental. Additionally, I will propose a 'tipping-point' model in which neutrophils are an integral part of a feedforward loop driving tuberculosis disease exacerbation., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
10. Loss of CD4 + T cell-intrinsic arginase 1 accelerates Th1 response kinetics and reduces lung pathology during influenza infection.
- Author
-
West EE, Merle NS, Kamiński MM, Palacios G, Kumar D, Wang L, Bibby JA, Overdahl K, Jarmusch AK, Freeley S, Lee DY, Thompson JW, Yu ZX, Taylor N, Sitbon M, Green DR, Bohrer A, Mayer-Barber KD, Afzali B, Kazemian M, Scholl-Buergi S, Karall D, Huemer M, and Kemper C
- Subjects
- Animals, Humans, Mice, CD4-Positive T-Lymphocytes metabolism, Glutamine, Kinetics, Lung metabolism, Mammals, Arginase genetics, Arginase metabolism, Influenza, Human
- Abstract
Arginase 1 (Arg1), the enzyme catalyzing the conversion of arginine to ornithine, is a hallmark of IL-10-producing immunoregulatory M2 macrophages. However, its expression in T cells is disputed. Here, we demonstrate that induction of Arg1 expression is a key feature of lung CD4
+ T cells during mouse in vivo influenza infection. Conditional ablation of Arg1 in CD4+ T cells accelerated both virus-specific T helper 1 (Th1) effector responses and its resolution, resulting in efficient viral clearance and reduced lung pathology. Using unbiased transcriptomics and metabolomics, we found that Arg1-deficiency was distinct from Arg2-deficiency and caused altered glutamine metabolism. Rebalancing this perturbed glutamine flux normalized the cellular Th1 response. CD4+ T cells from rare ARG1-deficient patients or CRISPR-Cas9-mediated ARG1-deletion in healthy donor cells phenocopied the murine cellular phenotype. Collectively, CD4+ T cell-intrinsic Arg1 functions as an unexpected rheostat regulating the kinetics of the mammalian Th1 lifecycle with implications for Th1-associated tissue pathologies., Competing Interests: Declaration of interests M.S. is an inventor on a patent describing the use of RBD ligands for cell-surface evaluation of CAT1/solute carrier family 7 member 1 (SLC7A1) and other solute carrier (SLC) expression (N.T. gave up her rights); he is a co-founder and head of the scientific board of METAFORA-Biosystems, a start-up company that focuses on metabolite transporters under physiological and pathological conditions., (Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
11. Co-infection of mice with SARS-CoV-2 and Mycobacterium tuberculosis limits early viral replication but does not affect mycobacterial loads.
- Author
-
Baker PJ, Amaral EP, Castro E, Bohrer AC, Torres-Juárez F, Jordan CM, Nelson CE, Barber DL, Johnson RF, Hilligan KL, and Mayer-Barber KD
- Subjects
- Mice, Animals, Humans, SARS-CoV-2, Pandemics, Mice, Transgenic, Mice, Inbred C57BL, Mycobacterium tuberculosis, Coinfection, COVID-19, Interferon Type I
- Abstract
Viral co-infections have been implicated in worsening tuberculosis (TB) and during the COVID-19 pandemic, the global rate of TB-related deaths has increased for the first time in over a decade. We and others have previously shown that a resolved prior or concurrent influenza A virus infection in Mycobacterium tuberculosis ( Mtb )-infected mice resulted in increased pulmonary bacterial burden, partly through type I interferon (IFN-I)-dependent mechanisms. Here we investigated whether SARS-CoV-2 (SCV2) co-infection could also negatively affect bacterial control of Mtb . Importantly, we found that K18-hACE2 transgenic mice infected with SCV2 one month before, or months after aerosol Mtb exposure did not display exacerbated Mtb infection-associated pathology, weight loss, nor did they have increased pulmonary bacterial loads. However, pre-existing Mtb infection at the time of exposure to the ancestral SCV2 strain in infected K18-hACE2 transgenic mice or the beta variant (B.1.351) in WT C57Bl/6 mice significantly limited early SCV2 replication in the lung. Mtb -driven protection against SCV2 increased with higher bacterial doses and did not require IFN-I, TLR2 or TLR9 signaling. These data suggest that SCV2 co-infection does not exacerbate Mtb infection in mice, but rather the inflammatory response generated by Mtb infection in the lungs at the time of SCV2 exposure restricts viral replication., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Baker, Amaral, Castro, Bohrer, Torres-Juárez, Jordan, Nelson, Barber, Johnson, Hilligan and Mayer-Barber.)
- Published
- 2023
- Full Text
- View/download PDF
12. Exposure to lung-migrating helminth protects against murine SARS-CoV-2 infection through macrophage-dependent T cell activation.
- Author
-
Oyesola OO, Hilligan KL, Namasivayam S, Howard N, Clancy CS, Zhao M, Oland SD, Kiwanuka KN, Garza NL, Lafont BAP, Johnson RF, Mayer-Barber KD, Sher A, and Loke P
- Subjects
- Mice, Humans, Animals, SARS-CoV-2, Macrophages, Lung, Mice, Transgenic, CD8-Positive T-Lymphocytes, COVID-19 metabolism
- Abstract
Helminth endemic regions report lower COVID-19 morbidity and mortality. Here, we show that lung remodeling from a prior infection with a lung-migrating helminth, Nippostrongylus brasiliensis , enhances viral clearance and survival of human-ACE2 transgenic mice challenged with SARS-CoV-2 (SCV2). This protection is associated with a lymphocytic infiltrate, including increased accumulation of pulmonary SCV2-specific CD8
+ T cells, and anti-CD8 antibody depletion abrogated the N. brasiliensis- mediated reduction in viral loads. Pulmonary macrophages with a type 2 transcriptional and epigenetic signature persist in the lungs of N. brasiliensis -exposed mice after clearance of the parasite and establish a primed environment for increased CD8+ T cell recruitment and activation. Accordingly, depletion of macrophages ablated the augmented viral clearance and accumulation of CD8+ T cells driven by prior N. brasiliensis infection. Together, these findings support the concept that lung-migrating helminths can limit disease severity during SCV2 infection through macrophage-dependent enhancement of antiviral CD8+ T cell responses.- Published
- 2023
- Full Text
- View/download PDF
13. A TNF-IL-1 circuit controls Yersinia within intestinal granulomas.
- Author
-
Matsuda R, Sorobetea D, Zhang J, Peterson ST, Grayczyk JP, Herrmann B, Yost W, O'Neill R, Bohrer AC, Lanza M, Assenmacher CA, Mayer-Barber KD, Shin S, and Brodsky IE
- Abstract
Tumor necrosis factor (TNF) is a pleiotropic inflammatory cytokine that mediates antimicrobial defense and granuloma formation in response to infection by numerous pathogens. Yersinia pseudotuberculosis colonizes the intestinal mucosa and induces recruitment of neutrophils and inflammatory monocytes into organized immune structures termed pyogranulomas that control the bacterial infection. Inflammatory monocytes are essential for control and clearance of Yersinia within intestinal pyogranulomas, but how monocytes mediate Yersinia restriction is poorly understood. Here, we demonstrate that TNF signaling in monocytes is required for bacterial containment following enteric Yersinia infection. We further show that monocyte-intrinsic TNFR1 signaling drives production of monocyte-derived interleukin-1 (IL-1), which signals through IL-1 receptor on non-hematopoietic cells to enable pyogranuloma-mediated control of Yersinia infection. Altogether, our work reveals a monocyte-intrinsic TNF-IL-1 collaborative circuit as a crucial driver of intestinal granuloma function, and defines the cellular target of TNF signaling that restricts intestinal Yersinia infection., Competing Interests: Competing interests The authors have no conflicting financial interests.
- Published
- 2023
- Full Text
- View/download PDF
14. Blood and Site of Disease Inflammatory Profiles Differ in Patients With Pericardial Tuberculosis and Human Immunodeficiency Virus Type 1.
- Author
-
Mutavhatsindi H, Du Bruyn E, Ruzive S, Howlett P, Cerrone M, Sher A, Mayer-Barber KD, Barber DL, Ntsekhe M, Wilkinson RJ, and Riou C
- Abstract
Background: To better understand the pathogenesis of pericardial tuberculosis (PCTB), we sought to characterize the systemic inflammatory profile in people with human immunodeficiency virus type 1 (HIV-1) with latent TB infection (LTBI), pulmonary TB (PTB), or PCTB., Methods: Using Luminex, we measured the concentration of 39 analytes in pericardial fluid (PCF) and paired plasma from 18 PCTB participants, and plasma from 16 LTBI and 20 PTB participants. Follow-up plasma samples were also obtained from PTB and PCTB participants. HLA-DR expression on Mycobacterium tuberculosis -specific CD4 T cells was measured in baseline samples using flow cytometry., Results: Assessment of the overall systemic inflammatory profile by principal component analysis showed that the inflammatory profile of active TB participants was distinct from the LTBI group, while PTB patients could not be distinguished from those with PCTB. When comparing the inflammatory profile between PCF and paired blood, we found that the concentrations of most analytes (25/39) were elevated at site of disease. However, the inflammatory profile in PCF partially mirrored inflammatory events in the blood. After TB treatment completion, the overall plasma inflammatory profile reverted to that observed in the LTBI group. Lastly, HLA-DR expression showed the best performance for TB diagnosis compared to previously described biosignatures built from soluble markers., Conclusions: Our results show that the inflammatory profile in blood was comparable between PTB and PCTB. However, at the site of infection (PCF), inflammation was significantly elevated compared to blood. Additionally, our data emphasize the potential role of HLA-DR expression as a biomarker for TB diagnosis., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)
- Published
- 2023
- Full Text
- View/download PDF
15. Corrigendum: Comparison of the frequency and phenotypic profile of Mycobacterium tuberculosis -specific CD4 T cells between the site of disease and blood in pericardial tuberculosis.
- Author
-
Du Bruyn E, Ruzive S, Howlett P, Cerrone M, Jacobs AJ, Lindestam Arlehamn CS, Sette A, Sher A, Mayer-Barber KD, Barber DL, Mayosi B, Ntsekhe M, Wilkinson RJ, and Riou C
- Abstract
[This corrects the article DOI: 10.3389/fimmu.2022.1009016.]., (Copyright © 2023 Du Bruyn, Ruzive, Howlett, Cerrone, Jacobs, Lindestam Arlehamn, Sette, Sher, Mayer-Barber, Barber, Mayosi, Ntsekhe, Wilkinson and Riou.)
- Published
- 2023
- Full Text
- View/download PDF
16. Human Dectin-1 deficiency impairs macrophage-mediated defense against phaeohyphomycosis.
- Author
-
Drummond RA, Desai JV, Hsu AP, Oikonomou V, Vinh DC, Acklin JA, Abers MS, Walkiewicz MA, Anzick SL, Swamydas M, Vautier S, Natarajan M, Oler AJ, Yamanaka D, Mayer-Barber KD, Iwakura Y, Bianchi D, Driscoll B, Hauck K, Kline A, Viall NS, Zerbe CS, Ferré EM, Schmitt MM, DiMaggio T, Pittaluga S, Butman JA, Zelazny AM, Shea YR, Arias CA, Ashbaugh C, Mahmood M, Temesgen Z, Theofiles AG, Nigo M, Moudgal V, Bloch KC, Kelly SG, Whitworth MS, Rao G, Whitener CJ, Mafi N, Gea-Banacloche J, Kenyon LC, Miller WR, Boggian K, Gilbert A, Sincock M, Freeman AF, Bennett JE, Hasbun R, Mikelis CM, Kwon-Chung KJ, Belkaid Y, Brown GD, Lim JK, Kuhns DB, Holland SM, and Lionakis MS
- Subjects
- Animals, Humans, Male, Mice, CARD Signaling Adaptor Proteins genetics, Lectins, C-Type genetics, Macrophages metabolism, Tumor Necrosis Factor-alpha genetics, beta-Glucans, Phaeohyphomycosis microbiology
- Abstract
Subcutaneous phaeohyphomycosis typically affects immunocompetent individuals following traumatic inoculation. Severe or disseminated infection can occur in CARD9 deficiency or after transplantation, but the mechanisms protecting against phaeohyphomycosis remain unclear. We evaluated a patient with progressive, refractory Corynespora cassiicola phaeohyphomycosis and found that he carried biallelic deleterious mutations in CLEC7A encoding the CARD9-coupled, β-glucan-binding receptor, Dectin-1. The patient's PBMCs failed to produce TNF-α and IL-1β in response to β-glucan and/or C. cassiicola. To confirm the cellular and molecular requirements for immunity against C. cassiicola, we developed a mouse model of this infection. Mouse macrophages required Dectin-1 and CARD9 for IL-1β and TNF-α production, which enhanced fungal killing in an interdependent manner. Deficiency of either Dectin-1 or CARD9 was associated with more severe fungal disease, recapitulating the human observation. Because these data implicated impaired Dectin-1 responses in susceptibility to phaeohyphomycosis, we evaluated 17 additional unrelated patients with severe forms of the infection. We found that 12 out of 17 carried deleterious CLEC7A mutations associated with an altered Dectin-1 extracellular C-terminal domain and impaired Dectin-1-dependent cytokine production. Thus, we show that Dectin-1 and CARD9 promote protective TNF-α- and IL-1β-mediated macrophage defense against C. cassiicola. More broadly, we demonstrate that human Dectin-1 deficiency may contribute to susceptibility to severe phaeohyphomycosis by certain dematiaceous fungi.
- Published
- 2022
- Full Text
- View/download PDF
17. Comparison of the frequency and phenotypic profile of Mycobacterium tuberculosis -specific CD4 T cells between the site of disease and blood in pericardial tuberculosis.
- Author
-
Du Bruyn E, Ruzive S, Howlett P, Jacobs AJ, Arlehamn CSL, Sette A, Sher A, Mayer-Barber KD, Barber DL, Mayosi B, Ntsekhe M, Wilkinson RJ, and Riou C
- Subjects
- Humans, CD4-Positive T-Lymphocytes, Interleukin-2 metabolism, Phenotype, Mycobacterium tuberculosis, Tuberculosis, Lymph Node
- Abstract
Studies of the immune response at the site of disease in extra-pulmonary tuberculosis (EPTB) disease are scarce. In this study, we compared the cellular profile of Mycobacterium tuberculosis (Mtb)-specific T cells in pericardial fluid and peripheral blood in patients with pericardial TB (PCTB). Whole blood and pericardial fluid (PCF) samples were collected at the time of diagnostic sampling, with repeat blood sampling after completion of anti-tubercular treatment (ATT) in 16 PCTB patients, most of them being HIV-1 infected (n=14). These samples were stimulated ex vivo and the phenotypic and functional cellular profile of PCF and blood was assessed by flow cytometry. We found that lymphocytes were the predominant cell type in PCF in PCTB, with a preferential influx of CD4 T cells. The frequencies of TNF-α producing Mtb-specific granulocytes and Mtb-specific CD4 T cells were significantly higher in PCF compared to blood. Mtb-specific CD4 T cells in PCF exhibited a distinct phenotype compared to those in blood, with greater GrB expression and lower CD27 and KLRG1 expression. We observed no difference in the production IFNγ, TNF or IL-2 by Mtb-specific CD4 T cells between the two compartments, but MIP-1β production was lower in the PCF T cells. Bacterial loads were not associated with alterations in the phenotype or function of Mtb-specific CD4 T cells. Upon ATT completion, HLA-DR, Ki-67 and GrB expression was significantly decreased, and relative IL-2 production was increased in peripheral Mtb-specific CD4 T cells. Overall, using an ex vivo assay to compare the immune response towards Mtb in PCF and in blood, we identified significant difference in the phenotypic profile of Mtb-specific CD4 T response between these two compartments. Moreover, we show that the activation profile of peripheral Mtb-specific CD4 T cells could be used to monitor treatment response in PCTB., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Du Bruyn, Ruzive, Howlett, Jacobs, Arlehamn, Sette, Sher, Mayer-Barber, Barber, Mayosi, Ntsekhe, Wilkinson and Riou.)
- Published
- 2022
- Full Text
- View/download PDF
18. Helminth exposure protects against murine SARS-CoV-2 infection through macrophage dependent T cell activation.
- Author
-
Hilligan KL, Oyesola OO, Namasivayam S, Howard N, Clancy CS, Oland SD, Garza NL, Lafont BAP, Johnson RF, Mayer-Barber KD, Sher A, and Loke P
- Abstract
Helminth endemic regions report lower COVID-19 morbidity and mortality. Here, we show that lung remodeling from a prior infection with a lung migrating helminth, Nippostrongylus brasiliensis , enhances viral clearance and survival of human-ACE2 transgenic mice challenged with SARS-CoV-2 (SCV2). This protection is associated with a lymphocytic infiltrate including an increased accumulation of pulmonary SCV2-specific CD8+ T cells and anti-CD8 antibody depletion abrogated the N. brasiliensis -mediated reduction in viral loads. Pulmonary macrophages with a type-2 transcriptional signature persist in the lungs of N. brasiliensis exposed mice after clearance of the parasite and establish a primed environment for increased antigen presentation. Accordingly, depletion of macrophages ablated the augmented viral clearance and accumulation of CD8+ T cells driven by prior N. brasiliensis infection. Together, these findings support the concept that lung migrating helminths can limit disease severity during SCV2 infection through macrophage-dependent enhancement of anti-viral CD8+ T cell responses.
- Published
- 2022
- Full Text
- View/download PDF
19. GPX4 regulates cellular necrosis and host resistance in Mycobacterium tuberculosis infection.
- Author
-
Amaral EP, Foreman TW, Namasivayam S, Hilligan KL, Kauffman KD, Barbosa Bomfim CC, Costa DL, Barreto-Duarte B, Gurgel-Rocha C, Santana MF, Cordeiro-Santos M, Du Bruyn E, Riou C, Aberman K, Wilkinson RJ, Barber DL, Mayer-Barber KD, Andrade BB, and Sher A
- Subjects
- Animals, Glutathione metabolism, Lipid Peroxidation, Mice, Mice, Transgenic, Necrosis, Phospholipid Hydroperoxide Glutathione Peroxidase, Ferroptosis, Glutathione Peroxidase metabolism, Tuberculosis immunology, Tuberculosis metabolism
- Abstract
Cellular necrosis during Mycobacterium tuberculosis (Mtb) infection promotes both immunopathology and bacterial dissemination. Glutathione peroxidase-4 (Gpx4) is an enzyme that plays a critical role in preventing iron-dependent lipid peroxidation-mediated cell death (ferroptosis), a process previously implicated in the necrotic pathology seen in Mtb-infected mice. Here, we document altered GPX4 expression, glutathione levels, and lipid peroxidation in patients with active tuberculosis and assess the role of this pathway in mice genetically deficient in or overexpressing Gpx4. We found that Gpx4-deficient mice infected with Mtb display substantially increased lung necrosis and bacterial burdens, while transgenic mice overexpressing the enzyme show decreased bacterial loads and necrosis. Moreover, Gpx4-deficient macrophages exhibited enhanced necrosis upon Mtb infection in vitro, an outcome suppressed by the lipid peroxidation inhibitor, ferrostatin-1. These findings provide support for the role of ferroptosis in Mtb-induced necrosis and implicate the Gpx4/GSH axis as a target for host-directed therapy of tuberculosis., (© 2022 Amaral et al.)
- Published
- 2022
- Full Text
- View/download PDF
20. Evaluation of endogenous and therapeutic 25-hydroxycholesterols in murine models of pulmonary SARS-CoV-2 infection.
- Author
-
Fessler MB, Madenspacher J, Baker PJ, Hilligan KL, Castro E, Meacham J, Chen SH, Johnson RF, Martin NP, Tucker CJ, Mahapatra D, Cesta M, and Mayer-Barber KD
- Abstract
Oxysterols (i.e., oxidized cholesterol species) have complex roles in biology. 25-hydroxycholesterol (25HC), a product of activity of cholesterol-25-hydroxylase (CH25H) upon cholesterol, has recently been shown to be broadly antiviral, suggesting therapeutic potential against SARS-CoV-2. However, 25HC can also amplify inflammation and tissue injury and be converted by CYP7B1 to 7α,25HC, a lipid with chemoattractant activity via the G protein-coupled receptor, EBI2/GPR183. Here, using in vitro studies and two different murine models of SARS-CoV-2 infection, we investigate the effects of these two oxysterols on SARS-CoV-2 pneumonia. We show that while 25HC and enantiomeric-25HC are antiviral in vitro against human endemic coronavirus-229E, they did not inhibit SARS-CoV-2; nor did supplemental 25HC reduce pulmonary SARS-CoV-2 titers in the K18-human ACE2 mouse model in vivo . 25HC treatment also did not alter immune cell influx into the airway, airspace cytokines, lung pathology, weight loss, symptoms, or survival but was associated with increased airspace albumin, an indicator of microvascular injury, and increased plasma pro-inflammatory cytokines. Conversely, mice treated with the EBI2/GPR183 inhibitor NIBR189 displayed a modest increase in lung viral load only at late time points, but no change in weight loss. Consistent with these findings, although Ch25h was upregulated in the lungs of SARS-CoV-2-infected WT mice, lung viral titers and weight loss in Ch25h
-/- and Gpr183-/- mice infected with the beta variant were similar to control animals. Taken together, endogenous 25-hydroxycholesterols do not significantly regulate early SARS-CoV-2 replication or pathogenesis and supplemental 25HC may have pro-injury rather than therapeutic effects in SARS-CoV-2 pneumonia.- Published
- 2022
- Full Text
- View/download PDF
21. Incidence of Nontuberculous Mycobacterial Pulmonary Infection, by Ethnic Group, Hawaii, USA, 2005-2019.
- Author
-
Blakney RA, Ricotta EE, Frankland TB, Honda S, Zelazny A, Mayer-Barber KD, Dean SG, Follmann D, Olivier KN, Daida YG, and Prevots DR
- Subjects
- Ethnicity, Hawaii epidemiology, Humans, Incidence, Nontuberculous Mycobacteria, Retrospective Studies, Mycobacterium Infections, Nontuberculous epidemiology, Mycobacterium Infections, Nontuberculous microbiology, Opportunistic Infections
- Abstract
To further clarify differences in the risk for nontuberculous mycobacterial pulmonary infection (NTM-PI) among ethnic populations in Hawaii, USA, we conducted a retrospective cohort study among beneficiaries of Kaiser Permanente Hawaii (KPH). We abstracted demographic, socioeconomic, clinical, and microbiological data from KPH electronic health records for 2005-2019. An NTM-PI case-patient was defined as a person from whom >1 NTM pulmonary isolate was obtained. We performed Cox proportional hazards regression to estimate incidence of NTM-PI while controlling for confounders. Across ethnic groups, risk for NTM-PI was higher among persons who were underweight (body mass index [BMI] <18.5 kg/m
2 ). Among beneficiaries who self-identified as any Asian ethnicity, risk for incident NTM-PI was increased by 30%. Low BMI may increase susceptibility to NTM-PI, and risk may be higher for persons who self-identify as Asian, independent of BMI.- Published
- 2022
- Full Text
- View/download PDF
22. MDA5 signaling induces type 1 IFN- and IL-1-dependent lung vascular permeability which protects mice from opportunistic fungal infection.
- Author
-
Davis MJ, Martin RE, Pinheiro GM, Hoke ES, Moyer S, Mayer-Barber KD, Chang YC, and Kwon-Chung KJ
- Subjects
- Animals, Capillary Permeability, Interleukin-1 metabolism, Interleukin-6 metabolism, Iron metabolism, Lung metabolism, Mice, Cryptococcosis metabolism, Cryptococcus gattii, Interferon Type I metabolism, Opportunistic Infections metabolism
- Abstract
Lungs balance threat from primary viral infection, secondary infection, and inflammatory damage. Severe pulmonary inflammation induces vascular permeability, edema, and organ dysfunction. We previously demonstrated that poly(I:C) (pICLC) induced type 1 interferon (t1IFN) protected mice from Cryptococcus gattii (Cg) via local iron restriction. Here we show pICLC increased serum protein and intravenously injected FITC-dextran in the lung airspace suggesting pICLC induces vascular permeability. Interestingly, pICLC induced a pro-inflammatory signature with significant expression of IL-1 and IL-6 which depended on MDA5 and t1IFN. Vascular permeability depended on MDA5, t1IFN, IL-1, and IL-6. T1IFN also induced MDA5 and other MDA5 signaling components suggesting that positive feedback contributes to t1IFN dependent expression of the pro-inflammatory signature. Vascular permeability, induced by pICLC or another compound, inhibited Cg by limiting iron. These data suggest that pICLC induces t1IFN which potentiates pICLC-MDA5 signaling increasing IL-1 and IL-6 resulting in leakage of antimicrobial serum factors into lung airspace. Thus, induced vascular permeability may act as an innate defense mechanism against opportunistic fungal infection, such as cryptococcosis, and may be exploited as a host-directed therapeutic target., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Davis, Martin, Pinheiro, Hoke, Moyer, Mayer-Barber, Chang and Kwon-Chung.)
- Published
- 2022
- Full Text
- View/download PDF
23. Rapid GPR183-mediated recruitment of eosinophils to the lung after Mycobacterium tuberculosis infection.
- Author
-
Bohrer AC, Castro E, Tocheny CE, Assmann M, Schwarz B, Bohrnsen E, Makiya MA, Legrand F, Hilligan KL, Baker PJ, Torres-Juarez F, Hu Z, Ma H, Wang L, Niu L, Wen Z, Lee SH, Kamenyeva O, Kauffman KD, Donato M, Sher A, Barber DL, Via LE, Scriba TJ, Khatri P, Song Y, Wong KW, Bosio CM, Klion AD, and Mayer-Barber KD
- Subjects
- Animals, Eosinophils metabolism, Humans, Lung pathology, Macrophages, Alveolar, Mice, Receptors, G-Protein-Coupled metabolism, Mycobacterium tuberculosis physiology, Tuberculosis pathology
- Abstract
Influx of eosinophils into the lungs is typically associated with type II responses during allergy and fungal and parasitic infections. However, we previously reported that eosinophils accumulate in lung lesions during type I inflammatory responses to Mycobacterium tuberculosis (Mtb) in humans, macaques, and mice, in which they support host resistance. Here we show eosinophils migrate into the lungs of macaques and mice as early as one week after Mtb exposure. In mice this influx is CCR3 independent and instead requires cell-intrinsic expression of the oxysterol receptor GPR183, which is highly expressed on human and macaque eosinophils. Murine eosinophils interact directly with bacilli-laden alveolar macrophages, which upregulate the oxysterol-synthesizing enzyme Ch25h, and eosinophil recruitment is impaired in Ch25h-deficient mice. Our findings show that eosinophils are among the earliest cells from circulation to sense and respond to Mtb infection of alveolar macrophages and reveal a role for GPR183 in the migration of eosinophils into lung tissue., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. ILC precursors differentiate into metabolically distinct ILC1-like cells during Mycobacterium tuberculosis infection.
- Author
-
Corral D, Charton A, Krauss MZ, Blanquart E, Levillain F, Lefrançais E, Sneperger T, Vahlas Z, Girard JP, Eberl G, Poquet Y, Guéry JC, Argüello RJ, Belkaid Y, Mayer-Barber KD, Hepworth MR, Neyrolles O, and Hudrisier D
- Subjects
- Cytokines, Humans, Inflammation, Lymphocytes, Immunity, Innate, Tuberculosis
- Abstract
Tissue-resident innate lymphoid cells (ILCs) regulate tissue homeostasis, protect against pathogens at mucosal surfaces, and are key players at the interface of innate and adaptive immunity. How ILCs adapt their phenotype and function to environmental cues within tissues remains to be fully understood. Here, we show that Mycobacterium tuberculosis (Mtb) infection alters the phenotype and function of lung IL-18Rα
+ ILC toward a protective interferon-γ-producing ILC1-like population. This differentiation is controlled by type 1 cytokines and is associated with a glycolytic program. Moreover, a BCG-driven type I milieu enhances the early generation of ILC1-like cells during secondary challenge with Mtb. Collectively, our data reveal how tissue-resident ILCs adapt to type 1 inflammation toward a pathogen-tailored immune response., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
25. Protein kinase R is an innate immune sensor of proteotoxic stress via accumulation of cytoplasmic IL-24.
- Author
-
Davidson S, Yu CH, Steiner A, Ebstein F, Baker PJ, Jarur-Chamy V, Hrovat Schaale K, Laohamonthonkul P, Kong K, Calleja DJ, Harapas CR, Balka KR, Mitchell J, Jackson JT, Geoghegan ND, Moghaddas F, Rogers KL, Mayer-Barber KD, De Jesus AA, De Nardo D, Kile BT, Sadler AJ, Poli MC, Krüger E, Goldbach Mansky R, and Masters SL
- Subjects
- Animals, Cells, Cultured, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, eIF-2 Kinase deficiency, Immunity, Innate immunology, Interleukins immunology, eIF-2 Kinase immunology
- Abstract
Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αβ) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.
- Published
- 2022
- Full Text
- View/download PDF
26. Intravenous administration of BCG protects mice against lethal SARS-CoV-2 challenge.
- Author
-
Hilligan KL, Namasivayam S, Clancy CS, O'Mard D, Oland SD, Robertson SJ, Baker PJ, Castro E, Garza NL, Lafont BAP, Johnson R, Ronchese F, Mayer-Barber KD, Best SM, and Sher A
- Subjects
- Administration, Intravenous, Angiotensin-Converting Enzyme 2 metabolism, Animals, Chemokines metabolism, Humans, Inflammation pathology, Mice, Inbred C57BL, Mice, Transgenic, Viral Load, Mice, BCG Vaccine immunology, COVID-19 immunology, SARS-CoV-2 immunology
- Abstract
In addition to providing partial protection against pediatric tuberculosis, vaccination with bacille Calmette-Guérin (BCG) has been reported to confer nonspecific resistance to unrelated pulmonary pathogens, a phenomenon attributed to the induction of long-lasting alterations within the myeloid cell compartment. Here, we demonstrate that intravenous, but not subcutaneous, inoculation of BCG protects human-ACE2 transgenic mice against lethal challenge with SARS-CoV-2 (SCV2) and results in reduced viral loads in non-transgenic animals infected with an α variant. The observed increase in host resistance was associated with reductions in SCV2-induced tissue pathology, inflammatory cell recruitment, and cytokine production that multivariate analysis revealed as only partially related to diminished viral load. We propose that this protection stems from BCG-induced alterations in the composition and function of the pulmonary cellular compartment that impact the innate response to the virus and ensuing immunopathology. While intravenous BCG vaccination is not a clinically acceptable practice, our findings provide an experimental model for identifying mechanisms by which nonspecific stimulation of the pulmonary immune response promotes host resistance to SCV2 lethality., Competing Interests: Disclosures: The authors declare no competing interests exist., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2022
- Full Text
- View/download PDF
27. Signature required: The transcriptional response to tuberculosis.
- Author
-
Barry CE and Mayer-Barber KD
- Subjects
- Gene Expression, Genetic Predisposition to Disease, Humans, Latent Tuberculosis genetics, Mycobacterium tuberculosis pathogenicity, Tuberculosis microbiology, Host-Pathogen Interactions physiology, Tuberculosis genetics, Tuberculosis transmission
- Abstract
The majority of humans infected with Mycobacterium tuberculosis never experience clinical symptoms or signs, but predicting those who will remains out of reach. Here, we discuss recent studies that reveal patterns and pathways that determine who is at highest risk for progression., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2021
- Full Text
- View/download PDF
28. Eosinophils are part of the granulocyte response in tuberculosis and promote host resistance in mice.
- Author
-
Bohrer AC, Castro E, Hu Z, Queiroz ATL, Tocheny CE, Assmann M, Sakai S, Nelson C, Baker PJ, Ma H, Wang L, Zilu W, du Bruyn E, Riou C, Kauffman KD, Moore IN, Del Nonno F, Petrone L, Goletti D, Martineau AR, Lowe DM, Cronan MR, Wilkinson RJ, Barry CE, Via LE, Barber DL, Klion AD, Andrade BB, Song Y, Wong KW, and Mayer-Barber KD
- Subjects
- Adult, Animals, Female, Granulocytes microbiology, Host-Pathogen Interactions physiology, Humans, Latent Tuberculosis microbiology, Lung pathology, Macaca mulatta, Male, Mice, Mutant Strains, Mycobacterium tuberculosis pathogenicity, Tuberculosis drug therapy, Zebrafish microbiology, Mice, Eosinophils physiology, Granulocytes physiology, Lung microbiology, Tuberculosis microbiology, Tuberculosis pathology
- Abstract
Host resistance to Mycobacterium tuberculosis (Mtb) infection requires the activities of multiple leukocyte subsets, yet the roles of the different innate effector cells during tuberculosis are incompletely understood. Here we uncover an unexpected association between eosinophils and Mtb infection. In humans, eosinophils are decreased in the blood but enriched in resected human tuberculosis lung lesions and autopsy granulomas. An influx of eosinophils is also evident in infected zebrafish, mice, and nonhuman primate granulomas, where they are functionally activated and degranulate. Importantly, using complementary genetic models of eosinophil deficiency, we demonstrate that in mice, eosinophils are required for optimal pulmonary bacterial control and host survival after Mtb infection. Collectively, our findings uncover an unexpected recruitment of eosinophils to the infected lung tissue and a protective role for these cells in the control of Mtb infection in mice., Competing Interests: Disclosures: D.M. Lowe reported personal fees from Merck, non-financial support from CSL Behring, and non-financial support from Fujifilm Chemical Co. outside the submitted work. No other disclosures were reported., (© 2021 Bohrer et al.)
- Published
- 2021
- Full Text
- View/download PDF
29. Intravenous administration of BCG protects mice against lethal SARS-CoV-2 challenge.
- Author
-
Hilligan KL, Namasivayam S, Clancy CS, O'Mard D, Oland SD, Robertson SJ, Baker PJ, Castro E, Garza NL, Lafont BAP, Johnson R, Ronchese F, Mayer-Barber KD, Best SM, and Sher A
- Abstract
Early events in the host response to SARS-CoV-2 are thought to play a major role in determining disease severity. During pulmonary infection, the virus encounters both myeloid and epithelioid lineage cells that can either support or restrict pathogen replication as well as respond with host protective versus detrimental mediators. In addition to providing partial protection against pediatric tuberculosis, vaccination with bacille Calmette-Guérin (BCG) has been reported to confer non-specific resistance to unrelated pulmonary pathogens, a phenomenon attributed to the induction of long-lasting alterations within the myeloid cell compartment. Here we demonstrate that prior intravenous, but not subcutaneous, administration of BCG protects human-ACE2 transgenic mice against lethal challenge with SARS-CoV-2 and results in reduced viral loads in non-transgenic animals infected with an alpha variant. The observed increase in host resistance was associated with reductions in SARS-CoV-2-induced tissue pathology, inflammatory cell recruitment and cytokine production that multivariate analysis revealed to be only partially related to diminished viral load. We propose that this protection stems from BCG-induced alterations in the composition and function of the pulmonary cellular compartment that impact the innate response to the virus and the ensuing immunopathology.
- Published
- 2021
- Full Text
- View/download PDF
30. Mycobacterium tuberculosis inhibits the NLRP3 inflammasome activation via its phosphokinase PknF.
- Author
-
Rastogi S, Ellinwood S, Augenstreich J, Mayer-Barber KD, and Briken V
- Subjects
- Animals, Host-Pathogen Interactions immunology, Mice, Mycobacterium tuberculosis metabolism, Protein Serine-Threonine Kinases metabolism, Tuberculosis metabolism, Immune Evasion immunology, Inflammasomes immunology, Mycobacterium tuberculosis immunology, NLR Family, Pyrin Domain-Containing 3 Protein immunology, Protein Serine-Threonine Kinases immunology, Tuberculosis immunology
- Abstract
Mycobacterium tuberculosis (Mtb) has evolved to evade host innate immunity by interfering with macrophage functions. Interleukin-1β (IL-1β) is secreted by macrophages after the activation of the inflammasome complex and is crucial for host defense against Mtb infections. We have previously shown that Mtb is able to inhibit activation of the AIM2 inflammasome and subsequent pyroptosis. Here we show that Mtb is also able to inhibit host cell NLRP3 inflammasome activation and pyroptosis. We identified the serine/threonine kinase PknF as one protein of Mtb involved in the NLRP3 inflammasome inhibition, since the pknF deletion mutant of Mtb induces increased production of IL-1β in bone marrow-derived macrophages (BMDMs). The increased production of IL-1β was dependent on NLRP3, the adaptor protein ASC and the protease caspase-1, as revealed by studies performed in gene-deficient BMDMs. Additionally, infection of BMDMs with the pknF deletion mutant resulted in increased pyroptosis, while the IL-6 production remained unchanged compared to Mtb-infected cells, suggesting that the mutant did not affect the priming step of inflammasome activation. In contrast, the activation step was affected since potassium efflux, chloride efflux and the generation of reactive oxygen species played a significant role in inflammasome activation and subsequent pyroptosis mediated by the Mtb pknF mutant strain. In conclusion, we reveal here that the serine/threonine kinase PknF of Mtb plays an important role in innate immune evasion through inhibition of the NLRP3 inflammasome., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
31. IL-1R1-Dependent Signals Improve Control of Cytosolic Virulent Mycobacteria In Vivo .
- Author
-
van der Niet S, van Zon M, de Punder K, Grootemaat A, Rutten S, Moorlag SJCFM, Houben D, van der Sar AM, Bitter W, Brosch R, Hernandez Pando R, Pena MT, Peters PJ, Reits EA, Mayer-Barber KD, and van der Wel NN
- Subjects
- Animals, Armadillos microbiology, Bacterial Translocation, Cytosol immunology, Female, Humans, Leprosy microbiology, Male, Mice, Mice, Inbred BALB C, Mice, SCID, Mycobacterium classification, Phagosomes immunology, Skin microbiology, Skin pathology, THP-1 Cells, Zebrafish, Cytosol microbiology, Mycobacterium immunology, Mycobacterium pathogenicity, Phagosomes microbiology, Receptors, Interleukin-1 genetics, Receptors, Interleukin-1 immunology, Signal Transduction immunology
- Abstract
Mycobacterium tuberculosis infections claim more than a million lives each year, and better treatments or vaccines are required. A crucial pathogenicity factor is translocation from phagolysosomes to the cytosol upon phagocytosis by macrophages. Translocation from the phagolysosome to the cytosol is an ESX-1-dependent process, as previously shown in vitro Here, we show that in vivo , mycobacteria also translocate to the cytosol but mainly when host immunity is compromised. We observed only low numbers of cytosolic bacilli in mice, armadillos, zebrafish, and patient material infected with M. tuberculosis , M. marinum , or M. leprae In contrast, when innate or adaptive immunity was compromised, as in severe combined immunodeficiency (SCID) or interleukin-1 receptor 1 (IL-1R1)-deficient mice, significant numbers of cytosolic M. tuberculosis bacilli were detected in the lungs of infected mice. Taken together, in vivo , translocation to the cytosol of M. tuberculosis is controlled by adaptive immune responses as well as IL-1R1-mediated signals. IMPORTANCE For decades, Mycobacterium tuberculosis has been one of the deadliest pathogens known. Despite infecting approximately one-third of the human population, no effective treatment or vaccine is available. A crucial pathogenicity factor is subcellular localization, as M. tuberculosis can translocate from phagolysosome to the cytosol in macrophages. The situation in vivo is more complicated. In this study, we establish that high-level cytosolic escape of mycobacteria can indeed occur in vivo but mainly when host resistance is compromised. The IL-1 pathway is crucial for the control of the number of cytosolic mycobacteria. The establishment that immune signals result in the clearance of cells containing cytosolic mycobacteria connects two important fields, cell biology and immunology, which is vital for the understanding of the pathology of M. tuberculosis ., (Copyright © 2021 van der Niet et al.)
- Published
- 2021
- Full Text
- View/download PDF
32. PD-1 blockade exacerbates Mycobacterium tuberculosis infection in rhesus macaques.
- Author
-
Kauffman KD, Sakai S, Lora NE, Namasivayam S, Baker PJ, Kamenyeva O, Foreman TW, Nelson CE, Oliveira-de-Souza D, Vinhaes CL, Yaniv Z, Lindestam Arleham CS, Sette A, Freeman GJ, Moore R, Sher A, Mayer-Barber KD, Andrade BB, Kabat J, Via LE, and Barber DL
- Subjects
- Animals, Bacterial Load drug effects, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CTLA-4 Antigen metabolism, Disease Models, Animal, Humans, Immune Checkpoint Inhibitors administration & dosage, Macaca mulatta, Male, Mice, Mice, Knockout, Mycobacterium tuberculosis immunology, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, Severity of Illness Index, Symptom Flare Up, Tuberculosis diagnosis, Tuberculosis immunology, Tuberculosis microbiology, CD4-Positive T-Lymphocytes drug effects, Immune Checkpoint Inhibitors adverse effects, Programmed Cell Death 1 Receptor antagonists & inhibitors, Tuberculosis drug therapy
- Abstract
Boosting immune cell function by targeting the coinhibitory receptor PD-1 may have applications in the treatment of chronic infections. Here, we examine the role of PD-1 during Mycobacterium tuberculosis (Mtb) infection of rhesus macaques. Animals treated with anti-PD-1 monoclonal antibody developed worse disease and higher granuloma bacterial loads compared with isotype control-treated monkeys. PD-1 blockade increased the number and functionality of granuloma Mtb-specific CD8 T cells. In contrast, Mtb-specific CD4 T cells in anti-PD-1-treated macaques were not increased in number or function in granulomas, expressed increased levels of CTLA-4, and exhibited reduced intralesional trafficking in live imaging studies. In granulomas of anti-PD-1-treated animals, multiple proinflammatory cytokines were elevated, and more cytokines correlated with bacterial loads, leading to the identification of a role for caspase 1 in the exacerbation of tuberculosis after PD-1 blockade. Last, increased Mtb bacterial loads after PD-1 blockade were found to associate with the composition of the intestinal microbiota before infection in individual macaques. Therefore, PD-1-mediated coinhibition is required for control of Mtb infection in macaques, perhaps because of its role in dampening detrimental inflammation and allowing for normal CD4 T cell responses., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
33. Development and Optimization of Chromosomally-Integrated Fluorescent Mycobacterium tuberculosis Reporter Constructs.
- Author
-
Kolbe K, Bell AC, Prosser GA, Assmann M, Yang HJ, Forbes HE, Gallucci S, Mayer-Barber KD, Boshoff HI, and Barry Iii CE
- Abstract
Mycobacterium tuberculosis resides in the lungs in various lesion types with unique microenvironmental conditions. This diversity is in line with heterogeneous disease progression and divergent drug efficiency. Fluorescent reporter strains can be used to decipher the micromilieu and to guide future treatment regimens. Current reporters using replicating plasmids, however, are not suitable for long-term mouse infections or studies in non-human primates. Using a combination of recombinant DNA and protein optimization techniques, we have developed reporter strains based on integrative plasmids, which exhibit stimulus-response characteristics and fluorescence intensities comparable to those based on replicating plasmids. We successfully applied the concepts by constructing a multi-color reporter strain able to detect simultaneous changes in environmental pH, Mg
2+ concentrations, and protein expression levels., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2020 Kolbe, Bell, Prosser, Assmann, Yang, Forbes, Gallucci, Mayer-Barber, Boshoff and Barry.)- Published
- 2020
- Full Text
- View/download PDF
34. Frontline Science: Anthrax lethal toxin-induced, NLRP1-mediated IL-1β release is a neutrophil and PAD4-dependent event.
- Author
-
Greaney AJ, Portley MK, O'Mard D, Crown D, Maier NK, Mendenhall MA, Mayer-Barber KD, Leppla SH, and Moayeri M
- Subjects
- Adaptor Proteins, Signal Transducing deficiency, Animals, Anthrax immunology, Antigens, Bacterial pharmacology, Apoptosis Regulatory Proteins deficiency, Bacillus anthracis physiology, Bacterial Toxins pharmacology, Inflammasomes physiology, Mice, Mice, 129 Strain, Mice, Congenic, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, SCID, Monocytes drug effects, Monocytes physiology, NLR Family, Pyrin Domain-Containing 3 Protein deficiency, Neutrophils drug effects, Protein-Arginine Deiminase Type 4 deficiency, Pyroptosis drug effects, Radiation Chimera, Species Specificity, Spores, Bacterial, Adaptor Proteins, Signal Transducing physiology, Antigens, Bacterial toxicity, Apoptosis Regulatory Proteins physiology, Bacillus anthracis pathogenicity, Bacterial Toxins toxicity, Extracellular Traps physiology, Interleukin-1beta metabolism, Neutrophils metabolism, Protein-Arginine Deiminase Type 4 physiology
- Abstract
Anthrax lethal toxin (LT) is a protease that activates the NLRP1b inflammasome sensor in certain rodent strains. Unlike better-studied sensors, relatively little is known about the priming requirements for NLRP1b. In this study, we investigate the rapid and striking priming-independent LT-induced release of IL-1β in mice within hours of toxin challenge. We find IL-1β release to be a NLRP1b- and caspase-1-dependent, NLRP3 and caspase-11-independent event that requires both neutrophils and peptidyl arginine deiminiase-4 (PAD4) activity. The simultaneous LT-induced IL-18 response is neutrophil-independent. Bone marrow reconstitution experiments in mice show toxin-induced IL-1β originates from hematopoietic cells. LT treatment of neutrophils in vitro did not induce IL-1β, neutrophil extracellular traps (NETs), or pyroptosis. Although platelets interact closely with neutrophils and are also a potential source of IL-1β, they were unable to bind or endocytose LT and did not secrete IL-1β in response to the toxin. LT-treated mice had higher levels of cell-free DNA and HMGB1 in circulation than PBS-treated controls, and treatment of mice with recombinant DNase reduced the neutrophil- and NLRP1-dependent IL-1β release. DNA sensor AIM2 deficiency, however, did not impact IL-1β release. These data, in combination with the findings on PAD4, suggest a possible role for in vivo NETs or cell-free DNA in cytokine induction in response to LT challenge. Our findings suggest a complex interaction of events and/or mediators in LT-treated mice with the neutrophil as a central player in induction of a profound and rapid inflammatory response to toxin., (©2020 Society for Leukocyte Biology.)
- Published
- 2020
- Full Text
- View/download PDF
35. Upregulation of CD47 Is a Host Checkpoint Response to Pathogen Recognition.
- Author
-
Tal MC, Torrez Dulgeroff LB, Myers L, Cham LB, Mayer-Barber KD, Bohrer AC, Castro E, Yiu YY, Lopez Angel C, Pham E, Carmody AB, Messer RJ, Gars E, Kortmann J, Markovic M, Hasenkrug M, Peterson KE, Winkler CW, Woods TA, Hansen P, Galloway S, Wagh D, Fram BJ, Nguyen T, Corey D, Kalluru RS, Banaei N, Rajadas J, Monack DM, Ahmed A, Sahoo D, Davis MM, Glenn JS, Adomati T, Lang KS, Weissman IL, and Hasenkrug KJ
- Subjects
- A549 Cells, Adaptive Immunity immunology, Animals, CD47 Antigen genetics, Cell Line, Tumor, Cytokines immunology, Female, Humans, Immunity, Innate immunology, Lymphocytic choriomeningitis virus immunology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mycobacterium tuberculosis immunology, SARS-CoV-2, Up-Regulation immunology, Betacoronavirus immunology, CD47 Antigen metabolism, Immunomodulation immunology, Receptors, Pattern Recognition immunology
- Abstract
It is well understood that the adaptive immune response to infectious agents includes a modulating suppressive component as well as an activating component. We now show that the very early innate response also has an immunosuppressive component. Infected cells upregulate the CD47 "don't eat me" signal, which slows the phagocytic uptake of dying and viable cells as well as downstream antigen-presenting cell (APC) functions. A CD47 mimic that acts as an essential virulence factor is encoded by all poxviruses, but CD47 expression on infected cells was found to be upregulated even by pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that encode no mimic. CD47 upregulation was revealed to be a host response induced by the stimulation of both endosomal and cytosolic pathogen recognition receptors (PRRs). Furthermore, proinflammatory cytokines, including those found in the plasma of hepatitis C patients, upregulated CD47 on uninfected dendritic cells, thereby linking innate modulation with downstream adaptive immune responses. Indeed, results from antibody-mediated CD47 blockade experiments as well as CD47 knockout mice revealed an immunosuppressive role for CD47 during infections with lymphocytic choriomeningitis virus and Mycobacterium tuberculosis Since CD47 blockade operates at the level of pattern recognition receptors rather than at a pathogen or antigen-specific level, these findings identify CD47 as a novel potential immunotherapeutic target for the enhancement of immune responses to a broad range of infectious agents. IMPORTANCE Immune responses to infectious agents are initiated when a pathogen or its components bind to pattern recognition receptors (PRRs). PRR binding sets off a cascade of events that activates immune responses. We now show that, in addition to activating immune responses, PRR signaling also initiates an immunosuppressive response, probably to limit inflammation. The importance of the current findings is that blockade of immunomodulatory signaling, which is mediated by the upregulation of the CD47 molecule, can lead to enhanced immune responses to any pathogen that triggers PRR signaling. Since most or all pathogens trigger PRRs, CD47 blockade could be used to speed up and strengthen both innate and adaptive immune responses when medically indicated. Such immunotherapy could be done without a requirement for knowing the HLA type of the individual, the specific antigens of the pathogen, or, in the case of bacterial infections, the antimicrobial resistance profile.
- Published
- 2020
- Full Text
- View/download PDF
36. Mouse transcriptome reveals potential signatures of protection and pathogenesis in human tuberculosis.
- Author
-
Moreira-Teixeira L, Tabone O, Graham CM, Singhania A, Stavropoulos E, Redford PS, Chakravarty P, Priestnall SL, Suarez-Bonnet A, Herbert E, Mayer-Barber KD, Sher A, Fonseca KL, Sousa J, Cá B, Verma R, Haldar P, Saraiva M, and O'Garra A
- Subjects
- Animals, B-Lymphocytes immunology, B-Lymphocytes microbiology, Humans, Interferon Type I immunology, Killer Cells, Natural immunology, Killer Cells, Natural microbiology, Lung immunology, Lung microbiology, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Mycobacterium tuberculosis immunology, T-Lymphocytes immunology, T-Lymphocytes microbiology, Tuberculosis microbiology, Transcriptome immunology, Tuberculosis immunology
- Abstract
Although mouse infection models have been extensively used to study the host response to Mycobacterium tuberculosis, their validity in revealing determinants of human tuberculosis (TB) resistance and disease progression has been heavily debated. Here, we show that the modular transcriptional signature in the blood of susceptible mice infected with a clinical isolate of M. tuberculosis resembles that of active human TB disease, with dominance of a type I interferon response and neutrophil activation and recruitment, together with a loss in B lymphocyte, natural killer and T cell effector responses. In addition, resistant but not susceptible strains of mice show increased lung B cell, natural killer and T cell effector responses in the lung upon infection. Notably, the blood signature of active disease shared by mice and humans is also evident in latent TB progressors before diagnosis, suggesting that these responses both predict and contribute to the pathogenesis of progressive M. tuberculosis infection.
- Published
- 2020
- Full Text
- View/download PDF
37. Diapedesis-Induced Integrin Signaling via LFA-1 Facilitates Tissue Immunity by Inducing Intrinsic Complement C3 Expression in Immune Cells.
- Author
-
Kolev M, West EE, Kunz N, Chauss D, Moseman EA, Rahman J, Freiwald T, Balmer ML, Lötscher J, Dimeloe S, Rosser EC, Wedderburn LR, Mayer-Barber KD, Bohrer A, Lavender P, Cope A, Wang L, Kaplan MJ, Moutsopoulos NM, McGavern D, Holland SM, Hess C, Kazemian M, Afzali B, and Kemper C
- Subjects
- Adult, Aged, Animals, Arthritis, Rheumatoid immunology, Arthritis, Rheumatoid metabolism, Arthritis, Rheumatoid pathology, Child, Child, Preschool, Complement C3 genetics, Complement C3 metabolism, Female, Humans, Integrins metabolism, Lymphocyte Function-Associated Antigen-1 metabolism, Lymphocytes metabolism, Male, Mice, Inbred C57BL, Middle Aged, Monocytes metabolism, Signal Transduction immunology, Complement C3 immunology, Integrins immunology, Lymphocyte Function-Associated Antigen-1 immunology, Lymphocytes immunology, Monocytes immunology, Transendothelial and Transepithelial Migration immunology
- Abstract
Intrinsic complement C3 activity is integral to human T helper type 1 (Th1) and cytotoxic T cell responses. Increased or decreased intracellular C3 results in autoimmunity and infections, respectively. The mechanisms regulating intracellular C3 expression remain undefined. We identified complement, including C3, as among the most significantly enriched biological pathway in tissue-occupying cells. We generated C3-reporter mice and confirmed that C3 expression was a defining feature of tissue-immune cells, including T cells and monocytes, occurred during transendothelial diapedesis, and depended on integrin lymphocyte-function-associated antigen 1 (LFA-1) signals. Immune cells from patients with leukocyte adhesion deficiency type 1 (LAD-1) had reduced C3 transcripts and diminished effector activities, which could be rescued proportionally by intracellular C3 provision. Conversely, increased C3 expression by T cells from arthritis patients correlated with disease severity. Our study defines integrins as key controllers of intracellular complement, demonstrates that perturbations in the LFA-1-C3-axis contribute to primary immunodeficiency, and identifies intracellular C3 as biomarker of severity in autoimmunity., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. Routemaps for Highly Effective Tuberculosis Vaccination.
- Author
-
Assmann M and Mayer-Barber KD
- Subjects
- Humans, Vaccination, Vaccines, Subunit, Mycobacterium tuberculosis immunology, Tuberculosis, Tuberculosis Vaccines
- Abstract
There is no highly effective tuberculosis vaccine. Darrah et al. (2020) and Tait et al. (2019) are setting new benchmarks for protection against infection and pulmonary disease by changing the route of vaccine delivery and by using a protein subunit vaccine with a potent adjuvant., (Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
39. Changes in inflammatory protein and lipid mediator profiles persist after antitubercular treatment of pulmonary and extrapulmonary tuberculosis: A prospective cohort study.
- Author
-
Vinhaes CL, Oliveira-de-Souza D, Silveira-Mattos PS, Nogueira B, Shi R, Wei W, Yuan X, Zhang G, Cai Y, Barry CE 3rd, Via LE, Fukutani KF, Andrade BB, and Mayer-Barber KD
- Subjects
- Adult, Biomarkers blood, Female, Humans, Male, Middle Aged, Prospective Studies, Antitubercular Agents administration & dosage, Cytokines blood, Cytokines immunology, Lipids blood, Lipids immunology, Mycobacterium tuberculosis immunology, Mycobacterium tuberculosis metabolism, Tuberculosis, Pulmonary blood, Tuberculosis, Pulmonary drug therapy, Tuberculosis, Pulmonary immunology
- Abstract
Background: The identification of meaningful biomarkers of tuberculosis (TB) has potential to improve diagnosis, disease staging and prediction of treatment outcomes. It has been shown that active pulmonary TB (PTB) is associated with qualitative and quantitative changes in systemic immune profile, suggesting a chronic inflammatory imbalance. Here we characterized the profile of PTB and extrapulmonary TB (EPTB) in a prospective cohort study., Methods: We measured a panel of 27 inflammatory cytokines, soluble receptors, and lipid mediators in peripheral blood from patients with PTB or EPTB from a prospective clinical study in China. Multidimensional analyses were performed to describe associations between plasma levels of biomarkers and different TB disease presentation profiles., Results: Mycobacterium tuberculosis infection induced changes in both the expression and correlation profiles of plasma mediators of inflammation in patients with PTB compared to those with EPTB. Increases in mycobacterial loads in sputum smears were associated with rises in concentrations of several molecules involved in TB pathogenesis, such as IL-1β, IFN-α, IL-10 and PGF2α. Moreover, PTB patients presenting with severe disease exhibited a distinct inflammatory profile hallmarked by heightened levels of TNF-α, IL-1β, IL17, IL-18 and IL-27. Interestingly, while antitubercular treatment (ATT) resulted in early changes of plasma concentrations of markers in PTB, changes were delayed in EPTB patients. Exploratory analyses of the molecular degree of perturbation (MDP) of the inflammatory mediators before and during ATT suggested the occurrence of infection and/or treatment-induced long lasting "inflammatory imprinting" of biomarker profiles in TB. At 24 weeks post ATT commencement, markers underlying the observed increases in MDP scores were IL-27 in PTB and IL-1β in EPTB patients., Conclusion: Our findings describe systemic and durable changes in the concentrations of inflammatory cytokines and lipid mediators in both PTB and EPTB and emphasize the role of M. tuberculosis bacterial burden and site of disease in modulating patient immune biomarkers., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
40. Targeting innate immunity for tuberculosis vaccination.
- Author
-
Khader SA, Divangahi M, Hanekom W, Hill PC, Maeurer M, Makar KW, Mayer-Barber KD, Mhlanga MM, Nemes E, Schlesinger LS, van Crevel R, Vankayalapati R(, Xavier RJ, and Netea MG
- Subjects
- Humans, BCG Vaccine immunology, BCG Vaccine therapeutic use, Immunity, Innate, Mycobacterium tuberculosis immunology, Tuberculosis immunology, Tuberculosis pathology, Tuberculosis prevention & control, Vaccination
- Abstract
Vaccine development against tuberculosis (TB) is based on the induction of adaptive immune responses endowed with long-term memory against mycobacterial antigens. Memory B and T cells initiate a rapid and robust immune response upon encounter with Mycobacterium tuberculosis, thus achieving long-lasting protection against infection. Recent studies have shown, however, that innate immune cell populations such as myeloid cells and NK cells also undergo functional adaptation after infection or vaccination, a de facto innate immune memory that is also termed trained immunity. Experimental and epidemiological data have shown that induction of trained immunity contributes to the beneficial heterologous effects of vaccines such as bacille Calmette-Guérin (BCG), the licensed TB vaccine. Moreover, increasing evidence argues that trained immunity also contributes to the anti-TB effects of BCG vaccination. An interaction among immunological signals, metabolic rewiring, and epigenetic reprogramming underlies the molecular mechanisms mediating trained immunity in myeloid cells and their bone marrow progenitors. Future studies are warranted to explore the untapped potential of trained immunity to develop a future generation of TB vaccines that would combine innate and adaptive immune memory induction.
- Published
- 2019
- Full Text
- View/download PDF
41. Transcriptional profiling unveils type I and II interferon networks in blood and tissues across diseases.
- Author
-
Singhania A, Graham CM, Gabryšová L, Moreira-Teixeira L, Stavropoulos E, Pitt JM, Chakravarty P, Warnatsch A, Branchett WJ, Conejero L, Lin JW, Davidson S, Wilson MS, Bancroft G, Langhorne J, Frickel E, Sesay AK, Priestnall SL, Herbert E, Ioannou M, Wang Q, Humphreys IR, Dodd J, Openshaw PJM, Mayer-Barber KD, Jankovic D, Sher A, Lloyd CM, Baldwin N, Chaussabel D, Papayannopoulos V, Wack A, Banchereau JF, Pascual VM, and O'Garra A
- Subjects
- Animals, Burkholderia pseudomallei, Candida albicans, Candidiasis immunology, Candidiasis microbiology, Gene Expression Regulation immunology, Influenza A Virus, H3N2 Subtype, Interferon Type I blood, Interferon Type I genetics, Interferon-gamma blood, Interferon-gamma genetics, Lung, Melioidosis immunology, Mice, Mice, Inbred C57BL, Orthomyxoviridae Infections genetics, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections virology, Receptor, Interferon alpha-beta, Receptors, Interferon, Respiratory Syncytial Virus Infections immunology, Interferon gamma Receptor, Candidiasis metabolism, Interferon Type I metabolism, Interferon-gamma metabolism, Melioidosis metabolism, Orthomyxoviridae Infections metabolism, Respiratory Syncytial Virus Infections metabolism
- Abstract
Understanding how immune challenges elicit different responses is critical for diagnosing and deciphering immune regulation. Using a modular strategy to interpret the complex transcriptional host response in mouse models of infection and inflammation, we show a breadth of immune responses in the lung. Lung immune signatures are dominated by either IFN-γ and IFN-inducible, IL-17-induced neutrophil- or allergy-associated gene expression. Type I IFN and IFN-γ-inducible, but not IL-17- or allergy-associated signatures, are preserved in the blood. While IL-17-associated genes identified in lung are detected in blood, the allergy signature is only detectable in blood CD4
+ effector cells. Type I IFN-inducible genes are abrogated in the absence of IFN-γ signaling and decrease in the absence of IFNAR signaling, both independently contributing to the regulation of granulocyte responses and pathology during Toxoplasma gondii infection. Our framework provides an ideal tool for comparative analyses of transcriptional signatures contributing to protection or pathogenesis in disease.- Published
- 2019
- Full Text
- View/download PDF
42. Pulmonary Iron Limitation Induced by Exogenous Type I IFN Protects Mice from Cryptococcus gattii Independently of T Cells.
- Author
-
Davis MJ, Moyer S, Hoke ES, Sionov E, Mayer-Barber KD, Barber DL, Cai H, Jenkins L, Walter PJ, Chang YC, and Kwon-Chung KJ
- Subjects
- Animals, Cryptococcus gattii drug effects, Disease Models, Animal, Female, Iron administration & dosage, Lung metabolism, Lung microbiology, Macrophages immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Poly I-C administration & dosage, Th1 Cells, Cryptococcosis immunology, Cryptococcosis prevention & control, Interferon Type I pharmacology, Iron metabolism, T-Lymphocytes immunology
- Abstract
Cryptococcus neoformans causes deadly mycosis primarily in AIDS patients, whereas Cryptococcus gattii infects mostly non-HIV patients, even in regions with high burdens of HIV/AIDS and an established environmental presence of C. gattii As HIV induces type I IFN (t1IFN), we hypothesized that t1IFN would differentially affect the outcome of C. neoformans and C. gattii infections. Exogenous t1IFN induction using stabilized poly(I·C) (pICLC) improved murine outcomes in either cryptococcal infection. In C. neoformans - infected mice, pICLC activity was associated with C. neoformans containment and classical Th1 immunity. In contrast, pICLC activity against C. gattii did not require any immune factors previously associated with C. neoformans immunity: T, B, and NK cells, IFN-γ, and macrophages were all dispensable. Interestingly, C. gattii pICLC activity depended on β-2-microglobulin, which impacts iron levels among other functions. Iron supplementation reversed pICLC activity, suggesting C. gattii pICLC activity requires iron limitation. Also, pICLC induced a set of iron control proteins, some of which were directly inhibitory to cryptococcus in vitro , suggesting t1IFN regulates iron availability in the pulmonary air space fluids. Thus, exogenous induction of t1IFN significantly improves the outcome of murine infection by C. gattii and C. neoformans but by distinct mechanisms; the C. gattii effect was mediated by iron limitation, while the effect on C. neoformans infection was through induction of classical T-cell-dependent immunity. Together this difference in types of T-cell-dependent t1IFN immunity for different Cryptococcus species suggests a possible mechanism by which HIV infection may select against C. gattii but not C. neoformans IMPORTANCE Cryptococcus neoformans and Cryptococcus gattii cause fatal infection in immunodeficient and immunocompetent individuals. While these fungi are sibling species, C. gattii infects very few AIDS patients, while C. neoformans infection is an AIDS-defining illness, suggesting that the host response to HIV selects C. neoformans over C. gattii We used a viral mimic molecule (pICLC) to stimulate the immune response, and pICLC treatment improved mouse outcomes from both species. pICLC-induced action against C. neoformans was due to activation of well-defined immune pathways known to deter C. neoformans , whereas these immune pathways were dispensable for pICLC treatment of C. gattii Since these immune pathways are eventually destroyed by HIV/AIDS, our data help explain why the antiviral immune response in AIDS patients is unable to control C. neoformans infection but is protective against C. gattii Furthermore, pICLC induced tighter control of iron in the lungs of mice, which inhibited C. gattii , thus suggesting an entirely new mode of nutritional immunity activated by viral signals.
- Published
- 2019
- Full Text
- View/download PDF
43. Molecular degree of perturbation of plasma inflammatory markers associated with tuberculosis reveals distinct disease profiles between Indian and Chinese populations.
- Author
-
Oliveira-de-Souza D, Vinhaes CL, Arriaga MB, Kumar NP, Cubillos-Angulo JM, Shi R, Wei W, Yuan X, Zhang G, Cai Y, Barry CE 3rd, Via LE, Sher A, Babu S, Mayer-Barber KD, Nakaya HI, Fukutani KF, and Andrade BB
- Subjects
- Adult, Blood Proteins genetics, China epidemiology, Cytokines blood, Female, Humans, India epidemiology, Inflammation blood, Inflammation microbiology, Inflammation pathology, Interferon-gamma blood, Latent Tuberculosis blood, Latent Tuberculosis microbiology, Latent Tuberculosis pathology, Lipids blood, Male, Mycobacterium tuberculosis pathogenicity, Tuberculosis, Pulmonary blood, Tuberculosis, Pulmonary microbiology, Tuberculosis, Pulmonary pathology, Tumor Necrosis Factor-alpha blood, Biomarkers blood, Inflammation epidemiology, Latent Tuberculosis epidemiology, Tuberculosis, Pulmonary epidemiology
- Abstract
Tuberculosis (TB) is a chronic inflammatory disease caused by Mycobacterium tuberculosis infection which causes tremendous morbidity and mortality worldwide. Clinical presentation of TB patients is very diverse and disease heterogeneity is associated with changes in biomarker signatures. Here, we compared at the molecular level the extent of individual inflammatory perturbation of plasma protein and lipid mediators associated with TB in patients in China versus India. We performed a cross-sectional study analyzing the overall degree of inflammatory perturbation in treatment-naïve pulmonary TB patients and uninfected individuals from India (TB: n = 97, healthy: n = 20) and China (TB: n = 100, healthy: n = 11). We employed the molecular degree of perturbation (MDP) adapted to plasma biomarkers to examine the overall changes in inflammation between these countries. M. tuberculosis infection caused a significant degree of molecular perturbation in patients from both countries, with higher perturbation detected in India. Interestingly, there were differences in biomarker perturbation patterns and the overall degree of inflammation. Patients with severe TB exhibited increased MDP values and Indian patients with this condition exhibited even higher degree of perturbation compared to Chinese patients. Network analyses identified IFN-α, IFN-β, IL-1RI and TNF-α as combined biomarkers that account for the overall molecular perturbation in the entire study population. Our results delineate the magnitude of the systemic inflammatory perturbation in pulmonary TB and reveal qualitative changes in inflammatory profiles between two countries with high disease prevalence.
- Published
- 2019
- Full Text
- View/download PDF
44. CARD9 + microglia promote antifungal immunity via IL-1β- and CXCL1-mediated neutrophil recruitment.
- Author
-
Drummond RA, Swamydas M, Oikonomou V, Zhai B, Dambuza IM, Schaefer BC, Bohrer AC, Mayer-Barber KD, Lira SA, Iwakura Y, Filler SG, Brown GD, Hube B, Naglik JR, Hohl TM, and Lionakis MS
- Subjects
- Animals, Brain immunology, Brain metabolism, Brain microbiology, CARD Signaling Adaptor Proteins genetics, CARD Signaling Adaptor Proteins metabolism, Candida albicans immunology, Candida albicans physiology, Candidiasis genetics, Candidiasis microbiology, Chemokine CXCL1 genetics, Chemokine CXCL1 metabolism, Cytokines genetics, Cytokines immunology, Cytokines metabolism, Host-Pathogen Interactions immunology, Inflammasomes genetics, Inflammasomes immunology, Inflammasomes metabolism, Interleukin-1beta genetics, Interleukin-1beta metabolism, Mice, Knockout, Mice, Transgenic, Microglia metabolism, Microglia microbiology, Neutrophil Infiltration genetics, Neutrophil Infiltration immunology, Neutrophils metabolism, Neutrophils microbiology, CARD Signaling Adaptor Proteins immunology, Candidiasis immunology, Chemokine CXCL1 immunology, Interleukin-1beta immunology, Microglia immunology, Neutrophils immunology
- Abstract
The C-type lectin receptor-Syk (spleen tyrosine kinase) adaptor CARD9 facilitates protective antifungal immunity within the central nervous system (CNS), as human deficiency in CARD9 causes susceptibility to fungus-specific, CNS-targeted infection. CARD9 promotes the recruitment of neutrophils to the fungus-infected CNS, which mediates fungal clearance. In the present study we investigated host and pathogen factors that promote protective neutrophil recruitment during invasion of the CNS by Candida albicans. The cytokine IL-1β served an essential function in CNS antifungal immunity by driving production of the chemokine CXCL1, which recruited neutrophils expressing the chemokine receptor CXCR2. Neutrophil-recruiting production of IL-1β and CXCL1 was induced in microglia by the fungus-secreted toxin Candidalysin, in a manner dependent on the kinase p38 and the transcription factor c-Fos. Notably, microglia relied on CARD9 for production of IL-1β, via both transcriptional regulation of Il1b and inflammasome activation, and of CXCL1 in the fungus-infected CNS. Microglia-specific Card9 deletion impaired the production of IL-1β and CXCL1 and neutrophil recruitment, and increased fungal proliferation in the CNS. Thus, an intricate network of host-pathogen interactions promotes antifungal immunity in the CNS; this is impaired in human deficiency in CARD9, which leads to fungal disease of the CNS.
- Published
- 2019
- Full Text
- View/download PDF
45. Mycobacterium tuberculosis Inhibits Autocrine Type I IFN Signaling to Increase Intracellular Survival.
- Author
-
Banks DA, Ahlbrand SE, Hughitt VK, Shah S, Mayer-Barber KD, Vogel SN, El-Sayed NM, and Briken V
- Subjects
- Animals, Autocrine Communication genetics, Interferon Type I genetics, Janus Kinase 1 genetics, Janus Kinase 1 immunology, Mice, Mice, Knockout, Microbial Viability genetics, Nitric Oxide genetics, Nitric Oxide immunology, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta immunology, Signal Transduction genetics, TYK2 Kinase genetics, TYK2 Kinase immunology, Autocrine Communication immunology, Interferon Type I immunology, Microbial Viability immunology, Mycobacterium tuberculosis immunology, Signal Transduction immunology
- Abstract
The type I IFNs (IFN-α and -β) are important for host defense against viral infections. In contrast, their role in defense against nonviral pathogens is more ambiguous. In this article, we report that IFN-β signaling in murine bone marrow-derived macrophages has a cell-intrinsic protective capacity against Mycobacterium tuberculosis via the increased production of NO. The antimycobacterial effects of type I IFNs were mediated by direct signaling through the IFN-α/β-receptor (IFNAR), as Ab-mediated blocking of IFNAR1 prevented the production of NO. Furthermore, M. tuberculosis is able to inhibit IFNAR-mediated cell signaling and the subsequent transcription of 309 IFN-β-stimulated genes in a dose-dependent way. The molecular mechanism of inhibition by M. tuberculosis involves reduced phosphorylation of the IFNAR-associated protein kinases JAK1 and TYK2, leading to reduced phosphorylation of the downstream targets STAT1 and STAT2. Transwell experiments demonstrated that the M. tuberculosis -mediated inhibition of type I IFN signaling was restricted to infected cells. Overall, our study supports the novel concept that M. tuberculosis evolved to inhibit autocrine type I IFN signaling to evade host defense mechanisms., (Copyright © 2019 by The American Association of Immunologists, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
46. A major role for ferroptosis in Mycobacterium tuberculosis -induced cell death and tissue necrosis.
- Author
-
Amaral EP, Costa DL, Namasivayam S, Riteau N, Kamenyeva O, Mittereder L, Mayer-Barber KD, Andrade BB, and Sher A
- Subjects
- Animals, Cell Death, Cells, Cultured, Ferroptosis drug effects, Glutathione metabolism, Host-Pathogen Interactions, Humans, Iron Chelating Agents pharmacology, Lipid Peroxidation, Macrophages drug effects, Macrophages microbiology, Macrophages pathology, Male, Mice, Inbred C57BL, Phospholipid Hydroperoxide Glutathione Peroxidase metabolism, Tuberculosis metabolism, Tuberculosis microbiology, Ferroptosis physiology, Iron metabolism, Mycobacterium tuberculosis pathogenicity, Tuberculosis pathology
- Abstract
Necrotic cell death during Mycobacterium tuberculosis (Mtb) infection is considered host detrimental since it facilitates mycobacterial spread. Ferroptosis is a type of regulated necrosis induced by accumulation of free iron and toxic lipid peroxides. We observed that Mtb-induced macrophage necrosis is associated with reduced levels of glutathione and glutathione peroxidase-4 (Gpx4), along with increased free iron, mitochondrial superoxide, and lipid peroxidation, all of which are important hallmarks of ferroptosis. Moreover, necrotic cell death in Mtb-infected macrophage cultures was suppressed by ferrostatin-1 (Fer-1), a well-characterized ferroptosis inhibitor, as well as by iron chelation. Additional experiments in vivo revealed that pulmonary necrosis in acutely infected mice is associated with reduced Gpx4 expression as well as increased lipid peroxidation and is likewise suppressed by Fer-1 treatment. Importantly, Fer-1-treated infected animals also exhibited marked reductions in bacterial load. Together, these findings implicate ferroptosis as a major mechanism of necrosis in Mtb infection and as a target for host-directed therapy of tuberculosis., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)
- Published
- 2019
- Full Text
- View/download PDF
47. Cutting Edge: IL-1R1 Mediates Host Resistance to Mycobacterium tuberculosis by Trans -Protection of Infected Cells.
- Author
-
Bohrer AC, Tocheny C, Assmann M, Ganusov VV, and Mayer-Barber KD
- Subjects
- Animals, Lung pathology, Macrophages immunology, Macrophages pathology, Mice, Mice, Knockout, Receptors, Interleukin-1 Type I genetics, Signal Transduction genetics, Tuberculosis, Pulmonary genetics, Tuberculosis, Pulmonary pathology, Immunity, Innate, Lung immunology, Mycobacterium tuberculosis immunology, Receptors, Interleukin-1 Type I immunology, Signal Transduction immunology, Tuberculosis, Pulmonary immunology
- Abstract
IL-1R1 deficiency in mice causes severe susceptibility to Mycobacterium tuberculosis Mice and macrophage cultures lacking IL-1R1 display increased bacterial growth, suggesting that phagocytes may require IL-1R1-dependent antimicrobial signals to limit intracellular M. tuberculosis replication directly. However, the myeloid-cell-intrinsic versus -extrinsic requirements for IL-1R1 to control M. tuberculosis infection in mice have not been directly addressed. Using single-cell analysis of infected cells, competitive mixed bone marrow chimeras, and IL-1R1 conditional mutant mice, we show in this article that IL-1R1 expression by pulmonary phagocytes is uncoupled from their ability to control intracellular M. tuberculosis growth. Importantly, IL-1R1-dependent control was provided to infected cells in trans by both nonhematopoietic and hematopoietic cells. Thus, IL-1R1-mediated host resistance to M. tuberculosis infection does not involve mechanisms of cell-autonomous antimicrobicidal effector functions in phagocytes but requires the cooperation between infected cells and other cells of hematopoietic or nonhematopoietic origin to promote bacterial containment and control of infection., (Copyright © 2018 by The American Association of Immunologists, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
48. Lysosomal Cathepsin Release Is Required for NLRP3-Inflammasome Activation by Mycobacterium tuberculosis in Infected Macrophages.
- Author
-
Amaral EP, Riteau N, Moayeri M, Maier N, Mayer-Barber KD, Pereira RM, Lage SL, Kubler A, Bishai WR, D'Império-Lima MR, Sher A, and Andrade BB
- Abstract
Lysosomal cathepsin B (CTSB) has been proposed to play a role in the induction of acute inflammation. We hypothesised that the presence of active CTSB in the cytosol is crucial for NLRP3-inflammasome assembly and, consequently, for mature IL-1β generation after mycobacterial infection in vitro . Elevated levels of CTSB was observed in the lungs of mice and rabbits following infection with Mycobacterium tuberculosis (Mtb) H37Rv as well as in plasma from acute tuberculosis patients. H37Rv-infected murine bone marrow-derived macrophages (BMDMs) displayed both lysosomal leakage, with release of CTSB into the cytosol, as well as increased levels of mature IL-1β. These responses were diminished in BMDM infected with a mutant H37Rv deficient in ESAT-6 expression. Pharmacological inhibition of cathepsin activity with CA074-Me resulted in a substantial reduction of both mature IL-1β production and caspase-1 activation in infected macrophages. Moreover, cathepsin inhibition abolished the interaction between NLRP3 and ASC, measured by immunofluorescence imaging in H37Rv-infected macrophages, demonstrating a critical role of the enzyme in NLRP3-inflammasome activation. These observations suggest that during Mtb infection, lysosomal release of activated CTSB and possibly other cathepsins inhibitable by CA07-Me is critical for the induction of inflammasome-mediated IL-1β processing by regulating NLRP3-inflammasome assembly in the cytosol.
- Published
- 2018
- Full Text
- View/download PDF
49. STAT-3-independent production of IL-17 by mouse innate-like αβ T cells controls ocular infection.
- Author
-
St Leger AJ, Hansen AM, Karauzum H, Horai R, Yu CR, Laurence A, Mayer-Barber KD, Silver P, Villasmil R, Egwuagu C, Datta SK, and Caspi RR
- Subjects
- Animals, Immunologic Memory, Interleukins metabolism, Mice, Inbred C57BL, Mice, Knockout, Mucous Membrane immunology, Mucous Membrane microbiology, Nuclear Receptor Subfamily 1, Group F, Member 3 metabolism, Phosphorylation, Promyelocytic Leukemia Zinc Finger Protein metabolism, Signal Transduction, Staphylococcus aureus physiology, T-Lymphocytes metabolism, Th17 Cells metabolism, Thymus Gland metabolism, Eye Infections immunology, Eye Infections pathology, Immunity, Innate, Interleukin-17 biosynthesis, Receptors, Antigen, T-Cell, alpha-beta metabolism, STAT3 Transcription Factor metabolism
- Abstract
Appropriate regulation of IL-17 production in the host can mean the difference between effective control of pathogens and uncontrolled inflammation that causes tissue damage. Investigation of conventional CD4
+ T cells (Th17 cells) has yielded invaluable insights into IL-17 function and its regulation. More recently, we and others reported production of IL-17 from innate αβ+ T cell populations, which was shown to occur primarily via IL-23R signaling through the transcription factor STAT-3. In our current study, we identify promyelocytic leukemia zinc finger (PLZF)-expressing iNKT, CD4- /CD8+ , and CD4- /CD8- (DN) αβ+T cells, which produce IL-17 in response to TCR and IL-1 receptor ligation independently of STAT-3 signaling. Notably, this noncanonical pathway of IL-17 production may be important in mucosal defense and is by itself sufficient to control pathogenic Staphylococcus aureus infection at the ocular surface., (This is a work of the U.S. Government and is not subject to copyright protection in the United States. Foreign copyrights may apply.)- Published
- 2018
- Full Text
- View/download PDF
50. Clash of the Cytokine Titans: counter-regulation of interleukin-1 and type I interferon-mediated inflammatory responses.
- Author
-
Mayer-Barber KD and Yan B
- Subjects
- Animals, Humans, Inflammation metabolism, Models, Biological, Cytokines metabolism, Inflammation pathology, Interferon Type I metabolism, Interleukin-1 metabolism
- Abstract
Over the past decades the notion of 'inflammation' has been extended beyond the original hallmarks of rubor (redness), calor (heat), tumor (swelling) and dolor (pain) described by Celsus. We have gained a more detailed understanding of the cellular players and molecular mediators of inflammation which is now being applied and extended to areas of biomedical research such as cancer, obesity, heart disease, metabolism, auto-inflammatory disorders, autoimmunity and infectious diseases. Innate cytokines are often central components of inflammatory responses. Here, we discuss how the type I interferon and interleukin-1 cytokine pathways represent distinct and specialized categories of inflammatory responses and how these key mediators of inflammation counter-regulate each other.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.