Neris RLS, Figueiredo CM, Higa LM, Araujo DF, Carvalho CAM, Verçoza BRF, Silva MOL, Carneiro FA, Tanuri A, Gomes AMO, Bozza MT, Da Poian AT, Cruz-Oliveira C, Assunção-Miranda I. Co-protoporphyrin IX and Sn-protoporphyrin IX inactivate Zika, Chikungunya and other arboviruses by targeting the viral envelope. Sci Rep. 2018 Jun 28;8(1):9805. 

DOI: 10.1038/s41598-018-27855-7

 ///

The global situation of diseases transmitted by arthropod-borne viruses such as Dengue (DENV), Yellow Fever (YFV), Chikungunya (CHIKV) and Zika (ZIKV) viruses is alarming and treatment of human infection by these arboviruses faces several challenges. The discovery of broad-spectrum antiviral molecules, able to inactivate different groups of viruses, is an interesting approach. The viral envelope is a common structure among arboviruses, being a potential target for antivirals. Porphyrins are amphipathic molecules able to interact with membranes and absorb light, being widely used in photodynamic therapy. Previously, we showed that heme, Co-protoporphyrin IX (CoPPIX) and Sn-protoporphyrin IX (SnPPIX) directly inactivate DENV and YFV infectious particles. Here we demonstrate that the antiviral activity of these porphyrins can be broadened to CHIKV, ZIKV, Mayaro virus, Sindbis virus and Vesicular Stomatitis virus. Porphyrin treatment causes viral envelope protein loss, affecting viral morphology, adsorption and entry into target cells. Also, light-stimulation enhanced the SnPPIX activity against all tested arboviruses. In summary, CoPPIX and SnPPIX were shown to be efficient broad-spectrum compounds to inactivate medically and veterinary important viruses.

Lucas CGO, Kitoko JZ, Ferreira FM, Suzart VG, Papa MP, Coelho SVA, Cavazzoni CB, Paula-Neto HA, Olsen PC, Iwasaki A, Pereira RM, Pimentel-Coelho PM, Vale AM, de Arruda LB, Bozza MT. Critical role of CD4+ T cells and IFNγ signaling in antibody-mediated resistance to Zika virus infection. Nat Commun. 2018 Aug 7;9(1):3136. 

DOI: 10.1038/s41467-018-05519-4 

///

Protective adaptive immunity to Zika virus (ZIKV) has been mainly attributed to cytotoxic CD8+ T cells and neutralizing antibodies, while the participation of CD4+ T cells in resistance has remained largely uncharacterized. Here, we show a neutralizing antibody response, dependent on CD4+ T cells and IFNγ signaling, which we detected during the first week of infection and is associated with reduced viral load in the brain, prevention of rapid disease onset and survival. We demonstrate participation of these components in the resistance to ZIKV during primary infection and in murine adoptive transfer models of heterologous ZIKV infection in a background of IFNR deficiency. The protective effect of adoptively transferred CD4+ T cells requires IFNγ signaling, CD8+ T cells and B lymphocytes in recipient mice. Together, this indicates the importance of CD4+ T cell responses in future vaccine design for ZIKV.

Abreu SC, Lopes-Pacheco M, da Silva AL, Xisto DG, de Oliveira TB, Kitoko JZ, de Castro LL, Amorim NR, Martins V, Silva LHA, Gonçalves-de-Albuquerque CF, de Castro Faria-Neto HC, Olsen PC, Weiss DJ, Morales MM, Diaz BL, Rocco PRM. Eicosapentaenoic Acid Enhances the Effects of Mesenchymal Stromal Cell Therapy in Experimental Allergic Asthma. Front Immunol. 2018 May 24;9:1147.

DOI: 10.3389/fimmu.2018.01147

///

Asthma is characterized by chronic lung inflammation and airway hyperresponsiveness. Despite recent advances in the understanding of its pathophysiology, asthma remains a major public health problem and, at present, there are no effective interventions capable of reversing airway remodeling. Mesenchymal stromal cell (MSC)-based therapy mitigates lung inflammation in experimental allergic asthma; however, its ability to reduce airway remodeling is limited. We aimed to investigate whether pre-treatment with eicosapentaenoic acid (EPA) potentiates the therapeutic properties of MSCs in experimental allergic asthma. Seventy-two C57BL/6 mice were used. House dust mite (HDM) extract was intranasally administered to induce severe allergic asthma in mice. Unstimulated or EPA-stimulated MSCs were administered intratracheally 24 h after final HDM challenge. Lung mechanics, histology, protein levels of biomarkers, and cellularity in bronchoalveolar lavage fluid (BALF), thymus, lymph nodes, and bone marrow were analyzed. Furthermore, the effects of EPA on lipid body formation and secretion of resolvin-D1 (RvD1), prostaglandin E2 (PGE2), interleukin (IL)-10, and transforming growth factor (TGF)-β1 by MSCs were evaluated in vitro. EPA-stimulated MSCs, compared to unstimulated MSCs, yielded greater therapeutic effects by further reducing bronchoconstriction, alveolar collapse, total cell counts (in BALF, bone marrow, and lymph nodes), and collagen fiber content in airways, while increasing IL-10 levels in BALF and M2 macrophage counts in lungs. In conclusion, EPA potentiated MSC-based therapy in experimental allergic asthma, leading to increased secretion of pro-resolution and anti-inflammatory mediators (RvD1, PGE2, IL-10, and TGF-β), modulation of macrophages toward an anti-inflammatory phenotype, and reduction in the remodeling process. Taken together, these modifications may explain the greater improvement in lung mechanics obtained. This may be a promising novel strategy to potentiate MSCs effects.

Muniz VS, Silva JC, Braga YAV, Melo RCN, Ueki S, Takeda M, Hebisawa A, Asano K, Figueiredo RT, Neves JS. Eosinophils release extracellular DNA traps in response to Aspergillus fumigatus. J Allergy Clin Immunol. 2018 Feb;141(2):571-585.e7. 

DOI: 10.1016/j.jaci.2017.07.048

///

Eosinophils mediate the immune response in different infectious conditions. The release of extracellular DNA traps (ETs) by leukocytes has been described as an innate immune response mechanism that is relevant in many disorders including fungal diseases. Different stimuli induce the release of human eosinophil ETs (EETs). Aspergillus fumigatus is an opportunistic fungus that may cause eosinophilic allergic bronchopulmonary aspergillosis (ABPA). It has been reported that eosinophils are important to the clearance of A fumigatus in infected mice lungs. However, the immunological mechanisms that underlie the molecular interactions between A fumigatus and eosinophils are poorly understood.

Banerjee I, Behl B, Mendonca M, Shrivastava G, Russo AJ, Menoret A, Ghosh A, Vella AT, Vanaja SK, Sarkar SN, Fitzgerald KA, Rathinam VAK. Gasdermin D Restrains Type I Interferon Response to Cytosolic DNA by Disrupting Ionic Homeostasis. Immunity. 2018 Sep 18;49(3):413-426.e5.

DOI: 10.1016/j.immuni.2018.07.006

///

Inflammasome complexes trigger the enzymatic activity of caspase-1, which activates interleukin-1β (IL-1β) and IL-18 (Martinon et al., 2002). Caspase-1 also targets gasdermin D, a pore-forming protein (Kayagaki et al., 2015, Shi et al., 2015). The pore-forming activity of gasdermin D resides in its N-terminal domain and is inhibited by its C-terminal domain. Caspase-1 cleaves gasdermin D at the linker region between these two domains, liberating the N-terminal domain, which migrates to the plasma membrane-forming pores with an inner diameter of 10–15 nm (Aglietti et al., 2016, Ding et al., 2016, Liu et al., 2016, Sborgi et al., 2016). An important consequence of gasdermin D activation is a lytic form of cell death called pyroptosis (He et al., 2015, Kayagaki et al., 2015, Shi et al., 2015). However, new evidence points out that gasdermin D executes additional functions independent of cell death; gasdermin D pores mediate the release of IL-1β and IL-18 without inducing cell death in response to certain ligands (Evavold et al., 2018). Additionally, after cytosolic LPS sensing by caspase-11, gasdermin D activates the NLRP3 inflammasome by inducing potassium (K+) efflux (Kayagaki et al., 2015, Rühl and Broz, 2015, Schmid-Burgk et al., 2015). Central to these distinct functions of gasdermin D is its membrane pore-forming activity. However, whether gasdermin D executes any additional immune functions is largely unknown.
A key surveillance mechanism in the cytosol, in addition to inflammasomes, is the cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) pathway. cGAS is a sensor for cytosolic DNA, and the binding of DNA by cGAS triggers its nucleotidyl transferase activity leading to the synthesis of cGAMP from ATP and GTP. cGAMP stimulates the transcription of type I interferon genes via the STING adaptor-TBK1 kinase-IRF3 transcription factor axis (Burdette et al., 2011, Wu et al., 2013). The type I interferon response elicited by cGAS plays important roles in host defense (Schneider et al., 2014). However, it is increasingly appreciated that the sustained production of type I interferons at high amounts is detrimental to the host, particularly during infections with intracellular bacteria such as Francisella novicida, Mycobacterium tuberculosis, and Listeria monocytogenes (Auerbuch et al., 2004, Henry et al., 2010, Mayer-Barber et al., 2014, McNab et al., 2015, Storek et al., 2015). Therefore, the magnitude and duration of cGAS-driven type I interferon responses should be kept in check. However, how the host restrains cGAS signaling during infections is poorly defined.
Here, we demonstrate that gasdermin D activated by the Aim2 inflammasome complex suppresses cytosolic DNA-induced production of type I interferons in macrophages. Consistent with this finding, mice lacking gasdermin D displayed enhanced IFN-β response to F. novicida infection. Pyroptosis and the extracellular release of IL-1 cytokines are dispensable for the inhibition of IFN-β by gasdermin D. Mechanistically, gasdermin D-induced membrane pores leaked intracellular potassium (K+) ions, and this K+ efflux in turn impaired type I interferon responses to cytosolic DNA and F. novicida. Gasdermin D-K+ efflux axis targeted cGAS to reduce cGAMP synthesis and thus IFN-β production. In summary, this study uncovers a previously unrecognized key role for gasdermin D in restraining cytosolic DNA-elicited interferon responses. Collectively, an emerging theme from the findings of this work and the recent studies (Evavold et al., 2018, Kayagaki et al., 2015) is that the fundamental pore-forming ability of gasdermin D and the consequent ionic fluxes confer gasdermin D additional biological functions independent of the terminal cell lytic event.

Gonçalves DS, Ferreira MDS, Liedke SC, Gomes KX, de Oliveira GA, Leão PEL, Cesar GV, Seabra SH, Cortines JR, Casadevall A, Nimrichter L, Domont GB, Junqueira MR, Peralta JM, Guimaraes AJ. Extracellular vesicles and vesicle-free secretome of the protozoa Acanthamoeba castellanii under homeostasis and nutritional stress and their damaging potential to host cells. Virulence. 2018 Dec 31;9(1):818-836.

DOI: 10.1080/21505594.2018.1451184

///

Acanthamoeba castellanii (Ac) are ubiquitously distributed in nature, and by contaminating medical devices such as heart valves and contact lenses, they cause a broad range of clinical presentations to humans. Although several molecules have been described to play a role in Ac pathogenesis, including parasite host-tissue invasion and escaping of host-defense, little information is available on their mechanisms of secretion. Herein, we describe the molecular components secreted by Ac, under different protein availability conditions to simulate host niches. Ac extracellular vesicles (EVs) were morphologically and biochemically characterized. Dynamic light scattering analysis of Ac EVs identified polydisperse populations, which correlated to electron microscopy measurements. High-performance thin liquid chromatography of Ac EVs identified phospholipids, steryl-esters, sterol and free-fatty acid, the last two also characterized by GC-MS. Secretome composition (EVs and EVs-free supernatants) was also determined and proteins biological functions classified. In peptone-yeast-glucose (PYG) medium, a total of 179 proteins were identified (21 common proteins, 89 exclusive of EVs and 69 in EVs-free supernatant). In glucose alone, 205 proteins were identified (134 in EVs, 14 common and 57 proteins in EVs-free supernatant). From those, stress response, oxidative and protein and amino acid metabolism proteins prevailed. Qualitative differences were observed on carbohydrate metabolism enzymes from Krebs cycle and pentose phosphate shunt. Serine proteases and metalloproteinases predominated. Analysis of the cytotoxicity of Ac EVs (upon uptake) and EVs-free supernatant to epithelial and glioblastoma cells revealed a dose-dependent effect. Therefore, the Ac secretome differs depending on nutrient conditions, and is also likely to vary during infection.

Vasconcellos LR, Siqueira MS, Moraes R, Carneiro LA, Bozza MT, Travassos LH. Heme Oxygenase-1 and Autophagy Linked for Cytoprotection. Curr Pharm Des. 2018;24(20):2311-2316. 

DOI: 10.2174/1381612824666180727100909

///

Heme-oxygenase (HO) catalyzes the main enzymatic step of heme degradation and generates anti-inflammatory end products with protective roles in physiological and pathological situations. The importance of HO in pathological conditions is evidenced by its pharmacological inhibition or genetic blockage in different models of stress such as infection, inflammation and oxidative stress. Under these situations, another well-known protective process triggered is autophagy. Autophagy is a homeostatic process that eliminates defective cytosolic components and organelles, allowing cells and tissues to recover through recycling of functional blocks for anabolic reactions. Recently, studies have demonstrated a link between HO activity and autophagy activation.

Topo