445 results on '"S. Zamboni"'
Search Results
2. Impact of histological variants in node positive patients treated with radical cystectomy for bladder cancer
- Author
-
M. Bandini, E. Zaffuto, S. Scuderi, G. Basile, A. Salonia, R. Lucianò, F. Pederzoli, S. Zamboni, L. Afferi, B. Giusy, M. Moschini, F. Dehò, R. Bertini, A. Briganti, F. Montorsi, R. Colombo, A. Necchi, and A. Gallina
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
3. No-response at mpMRI after neoadjuvant pembrolizumab is a proxy of adverse pathological and oncological outcomes in patients treated with radical cystectomy: interim results from the PURE01 study
- Author
-
G. Basile, M. Bandini, E. Zaffuto, S. Scuderi, F. Pederzoli, L. Marandino, D. Raggi, F. Barletta, G. Gandaglia, N. Fossati, G. Burgio, M. Moschini, S. Zamboni, L. Afferi, S. Comana, A. Briganti, F. Montorsi, R. Colombo, A. Necchi, and A. Gallina
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
4. Limited vs extended lymph node dissection in T1G3 bladder cancer patients treated with radical cystectomy: is this group responsible for the negative results of the LEA trial?
- Author
-
G. Basile, M. Bandini, E. Zaffuto, S. Scuderi, F. Dehò, A. Salonia, R. Matloob, G. Burgio, L. Sciacqua, R. Bertini, S. Comana, M. Moschini, S. Zamboni, L. Afferi, V. Scattoni, R. Colombo, A. Briganti, F. Montorsi, A. Necchi, and A. Gallina
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. No-response at mpMRI after neoadjuvant pembrolizumab is a proxy of adverse pathological and oncological outcomes in patients treated with radical cystectomy: Interim results from the PURE01 study
- Author
-
M. Bandini, E. Zaffuto, S. Scuderi, F. Pederzoli, L. Marandino, D. Raggi, F. Barletta, G. Gandaglia, N. Fossati, G. Burgio, M. Moschini, S. Zamboni, L. Afferi, S. Comara, A. Briganti, F. Montorsi, R. Colombo, A. Necchi, and A. Gallina
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
6. No impact on disease recurrence or progression of the stage at re-TUR in Ta high-grade bladder cancer patients treated with BCG: A multi-center collaboration of EAU Young Academic Urologists Urothelial Carcinoma Group
- Author
-
S. Régnier, M. Moschini, F. Soria, J. Dobruch, D. D`andrea, S.F. Shariat, A. Budowski, C. Poyet, M. Roumiguie, F.X. Nouhaud, M. Alvarez Maestro, A. Briganti, F. Montorsi, W. Krajewski, K. Hendricksen, H. Veerman, L. Afferi, A. Mattei, C. Di Bona, S. Zamboni, C. Simeone, A. Aziz, G. Verhoest, R. Thenault, and E. Xylinas
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
7. Adjuvant chemotherapy is ineffective in patients with bladder cancer and variant histology treated with radical cystectomy
- Author
-
S. Zamboni, P. Baumeister, A. Aziz, C. Poyet, C. Simeone, A. Antonelli, F. Roghmann, A. Meraney, B.R. Konety, F. Montorsi, A. Gallina, A. Briganti, G. Gandaglia, E. Zaffuto, G. Simone, R.J. Karnes, K. Hendricksen, C.S. Voskuilen, A. Mattei, L. Afferi, M. Rink, A. Necchi, S.F. Shariat, E. Xylinas, and M. Moschini
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
8. Mechanisms regulating host cell death during Leishmania infection
- Author
-
Juliane C. R. Fernandes and Dario S. Zamboni
- Subjects
Leishmania ,macrophages ,cell death ,apoptosis ,pyroptosis ,Microbiology ,QR1-502 - Abstract
ABSTRACT Parasites from the Leishmania genus are the causative agents of leishmaniasis and primarily reside within macrophages during mammalian infection. Their ability to establish intracellular infection provides a secure niche for proliferation while evading detection. However, successful multiplication within mammalian cells requires the orchestration of multiple mechanisms that control host cell viability. In contrast, innate immune cells, such as macrophages, can undergo different forms of cell death in response to pathogenic intracellular microbes. Thus, modulation of these different forms of host cell death is crucial for Leishmaniasis development. The regulation of host cell apoptosis, a form of programmed cell death, is crucial for sustaining parasites within viable host cells. Accordingly, several studies have demonstrated evasion of apoptosis induced by dermotropic and viscerotropic Leishmania species. Conversely, the prevention of pyroptosis, an inflammatory form of cell death, ensures the establishment of infection by silencing the release of mediators that could trigger massive proinflammatory responses. This manuscript explores how Leishmania regulates various host cell death pathways and overviews seminal studies on regulating host cell apoptosis by different Leishmania species.
- Published
- 2024
- Full Text
- View/download PDF
9. Pulmonary inflammation and viral replication define distinct clinical outcomes in fatal cases of COVID-19.
- Author
-
Keyla S G de Sá, Luana A Amaral, Tamara S Rodrigues, Camila C S Caetano, Amanda Becerra, Sabrina S Batah, Felipe T Lopes, Isadora M de Oliveira, Letícia S Lopes, Leticia Almeida, Caroline M Mota, Samuel Oliveira, Danilo T Wada, Marcel Koenigkam-Santos, Ronaldo B Martins, Roberta R C Rosales, Eurico Arruda, Alexandre T Fabro, and Dario S Zamboni
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
COVID-19 has affected more than half a billion people worldwide, with more than 6.3 million deaths, but the pathophysiological mechanisms involved in lethal cases and the host determinants that determine the different clinical outcomes are still unclear. In this study, we assessed lung autopsies of 47 COVID-19 patients and examined the inflammatory profiles, viral loads, and inflammasome activation. Additionally, we correlated these factors with the patient's clinical and histopathological conditions. Robust inflammasome activation was detected in the lungs of lethal cases of SARS-CoV-2. Experiments conducted on transgenic mice expressing hACE2 and infected with SARS-CoV-2 showed that Nlrp3-/- mice were protected from disease development and lethality compared to Nlrp3+/+ littermate mice, supporting the involvement of this inflammasome in disease exacerbation. An analysis of gene expression allowed for the classification of COVID-19 patients into two different clusters. Cluster 1 died with higher viral loads and exhibited a reduced inflammatory profile than Cluster 2. Illness time, mechanical ventilation time, pulmonary fibrosis, respiratory functions, histopathological status, thrombosis, viral loads, and inflammasome activation significantly differed between the two clusters. Our data demonstrated two distinct profiles in lethal cases of COVID-19, thus indicating that the balance of viral replication and inflammasome-mediated pulmonary inflammation led to different clinical outcomes. We provide important information to understand clinical variations in severe COVID-19, a process that is critical for decisions between immune-mediated or antiviral-mediated therapies for the treatment of critical cases of COVID-19.
- Published
- 2024
- Full Text
- View/download PDF
10. AIM2 promotes TH17 cells differentiation by regulating RORγt transcription activity
- Author
-
Jefferson Antônio Leite, Luísa Menezes, Eloisa Martins, Tamara Silva Rodrigues, Lucas Tavares, Anna Ebering, Carsten Schelmbauer, Guilherme C. Martelossi Cebinelli, Valeriya Zinina, Artemiy Golden, Natalia Soshnikova, Dario S. Zamboni, Fernando Q. Cunha, Magdalena Huber, João Santana Silva, Ari Waisman, Daniela Carlos, and Niels Olsen Saraiva Câmara
- Subjects
Biological sciences ,Cell biology ,Science - Abstract
Summary: AIM2 is an interferon-inducible HIN-200 protein family member and is well-documented for its roles in innate immune responses as a DNA sensor. Recent studies have highlighted AIM2’s function on regulatory T cells (Treg) and follicular T cells (Tfh). However, its involvement in Th17 cell differentiation remains unclear. This study reveals that AIM2 promotes Th17 cell differentiation. AIM2 deficiency decreases IL-17A production and downregulates key Th17 associated proteins (RORγt, IL-1R1, IL-23R). AIM2 is located in the nucleus of Th17 cells, where it interacts with RORγt, enhancing its binding to the Il17a promoter. The absence of AIM2 hinders naive CD4 T cells from differentiating into functional Th17 cells and from inducing colitis in Rag1−/− mice. This study uncovers AIM2’s role as a regulator of Th17 cell transcriptional programming, highlighting its potential as a therapeutic target for Th17 cell-mediated inflammatory diseases.
- Published
- 2023
- Full Text
- View/download PDF
11. Gasdermin-D activation promotes NLRP3 activation and host resistance to Leishmania infection
- Author
-
Keyla S. G. de Sá, Luana A. Amaral, Tamara S. Rodrigues, Adriene Y. Ishimoto, Warrison A. C. de Andrade, Leticia de Almeida, Felipe Freitas-Castro, Sabrina S. Batah, Sergio C. Oliveira, Mônica T. Pastorello, Alexandre T. Fabro, and Dario S. Zamboni
- Subjects
Science - Abstract
Here, de Sá et al. show that Gasdermin-D is transiently activated in Leishmania-infected macrophages and promotes NLRP3 inflammasome activation, but not cell death. Gasdermin-D is cleaved into a noncanonical fragment, indicating that Leishmania subverts Gasdermin-D-mediated host response to establish leishmaniasis.
- Published
- 2023
- Full Text
- View/download PDF
12. C5aR1 signaling triggers lung immunopathology in COVID-19 through neutrophil extracellular traps
- Author
-
Bruna M. Silva, Giovanni F. Gomes, Flavio P. Veras, Seppe Cambier, Gabriel V.L. Silva, Andreza U. Quadros, Diego B. Caetité, Daniele C. Nascimento, Camilla M. Silva, Juliana C. Silva, Samara Damasceno, Ayda H. Schneider, Fabio Beretta, Sabrina S. Batah, Icaro M.S. Castro, Isadora M. Paiva, Tamara Rodrigues, Ana Salina, Ronaldo Martins, Guilherme C.M. Cebinelli, Naira L. Bibo, Daniel M. Jorge, Helder I. Nakaya, Dario S. Zamboni, Luiz O. Leiria, Alexandre T. Fabro, José C. Alves-Filho, Eurico Arruda, Paulo Louzada-Junior, Renê D. Oliveira, Larissa D. Cunha, Pierre Van Mol, Lore Vanderbeke, Simon Feys, Els Wauters, Laura Brandolini, Andrea Aramini, Fernando Q. Cunha, Jörg Köhl, Marcello Allegretti, Diether Lambrechts, Joost Wauters, Paul Proost, and Thiago M. Cunha
- Subjects
COVID-19 ,Inflammation ,Medicine - Abstract
Patients with severe COVID-19 develop acute respiratory distress syndrome (ARDS) that may progress to cytokine storm syndrome, organ dysfunction, and death. Considering that complement component 5a (C5a), through its cellular receptor C5aR1, has potent proinflammatory actions and plays immunopathological roles in inflammatory diseases, we investigated whether the C5a/C5aR1 pathway could be involved in COVID-19 pathophysiology. C5a/C5aR1 signaling increased locally in the lung, especially in neutrophils of critically ill patients with COVID-19 compared with patients with influenza infection, as well as in the lung tissue of K18-hACE2 Tg mice (Tg mice) infected with SARS-CoV-2. Genetic and pharmacological inhibition of C5aR1 signaling ameliorated lung immunopathology in Tg-infected mice. Mechanistically, we found that C5aR1 signaling drives neutrophil extracellular traps-dependent (NETs-dependent) immunopathology. These data confirm the immunopathological role of C5a/C5aR1 signaling in COVID-19 and indicate that antagonists of C5aR1 could be useful for COVID-19 treatment.
- Published
- 2023
- Full Text
- View/download PDF
13. Gasdermin-D activation by SARS-CoV-2 triggers NET and mediate COVID-19 immunopathology
- Author
-
Camila Meirelles S. Silva, Carlos Wagner S. Wanderley, Flavio Protasio Veras, Augusto Velozo Gonçalves, Mikhael Haruo Fernandes Lima, Juliana Escher Toller-Kawahisa, Giovanni Freitas Gomes, Daniele Carvalho Nascimento, Valter V. Silva Monteiro, Isadora Marques Paiva, Cícero José Luíz Ramos Almeida, Diego Brito Caetité, Juliana Costa Silva, Maria Isabel Fernandes Lopes, Letícia Pastorelli Bonjorno, Marcela Cavichioli Giannini, Natalia Brasil Amaral, Maíra Nilson Benatti, Rodrigo Carvalho Santana, Luis Eduardo Alves Damasceno, Bruna Manuella Souza Silva, Ayda Henriques Schneider, Icaro Maia Santos Castro, Juan Carlo Santos Silva, Amanda Pereira Vasconcelos, Tiago Tomazini Gonçalves, Sabrina Setembre Batah, Tamara Silva Rodrigues, Victor Ferreira Costa, Marjorie Cornejo Pontelli, Ronaldo B. Martins, Timna Varela Martins, Danillo Lucas Alves Espósito, Guilherme Cesar Martelossi Cebinelli, Benedito Antônio Lopes da Fonseca, Luiz Osório Silveira Leiria, Larissa Dias Cunha, Eurico Arruda, Helder I. Nakaia, Alexandre Todorovic Fabro, Rene D. R. Oliveira, Dario S. Zamboni, Paulo Louzada-Junior, Thiago Mattar Cunha, José Carlos Farias Alves-Filho, and Fernando Queiroz Cunha
- Subjects
Neutrophil ,Innate immunity ,Organ damage ,COVID-19 ,NETs ,Medical emergencies. Critical care. Intensive care. First aid ,RC86-88.9 - Abstract
Abstract Background The release of neutrophil extracellular traps (NETs) is associated with inflammation, coagulopathy, and organ damage found in severe cases of COVID-19. However, the molecular mechanisms underlying the release of NETs in COVID-19 remain unclear. Objectives We aim to investigate the role of the Gasdermin-D (GSDMD) pathway on NETs release and the development of organ damage during COVID-19. Methods We performed a single-cell transcriptome analysis in public data of bronchoalveolar lavage. Then, we enrolled 63 hospitalized patients with moderate and severe COVID-19. We analyze in blood and lung tissue samples the expression of GSDMD, presence of NETs, and signaling pathways upstreaming. Furthermore, we analyzed the treatment with disulfiram in a mouse model of SARS-CoV-2 infection. Results We found that the SARS-CoV-2 virus directly activates the pore-forming protein GSDMD that triggers NET production and organ damage in COVID-19. Single-cell transcriptome analysis revealed that the expression of GSDMD and inflammasome-related genes were increased in COVID-19 patients. High expression of active GSDMD associated with NETs structures was found in the lung tissue of COVID-19 patients. Furthermore, we showed that activation of GSDMD in neutrophils requires active caspase1/4 and live SARS-CoV-2, which infects neutrophils. In a mouse model of SARS-CoV-2 infection, the treatment with disulfiram inhibited NETs release and reduced organ damage. Conclusion These results demonstrated that GSDMD-dependent NETosis plays a critical role in COVID-19 immunopathology and suggests GSDMD as a novel potential target for improving the COVID-19 therapeutic strategy.
- Published
- 2022
- Full Text
- View/download PDF
14. Efferocytosis of SARS-CoV-2-infected dying cells impairs macrophage anti-inflammatory functions and clearance of apoptotic cells
- Author
-
Ana CG Salina, Douglas dos-Santos, Tamara S Rodrigues, Marlon Fortes-Rocha, Edismauro G Freitas-Filho, Daniel L Alzamora-Terrel, Icaro MS Castro, Thais FC Fraga da Silva, Mikhael HF de Lima, Daniele C Nascimento, Camila M Silva, Juliana E Toller-Kawahisa, Amanda Becerra, Samuel Oliveira, Diego B Caetité, Leticia Almeida, Adriene Y Ishimoto, Thais M Lima, Ronaldo B Martins, Flavio Veras, Natália B do Amaral, Marcela C Giannini, Letícia P Bonjorno, Maria IF Lopes, Maira N Benatti, Sabrina S Batah, Rodrigo C Santana, Fernando C Vilar, Maria A Martins, Rodrigo L Assad, Sergio CL de Almeida, Fabiola R de Oliveira, Eurico Arruda Neto, Thiago M Cunha, José C Alves-Filho, Vania LD Bonato, Fernando Q Cunha, Alexandre T Fabro, Helder I Nakaya, Dario S Zamboni, Paulo Louzada-Junior, Rene DR Oliveira, and Larissa D Cunha
- Subjects
COVID-19 ,SARS-CoV-2 ,macrophage polarization ,efferocytosis ,hyperinflammation ,tissue repair ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
COVID-19 is a disease of dysfunctional immune responses, but the mechanisms triggering immunopathogenesis are not established. The functional plasticity of macrophages allows this cell type to promote pathogen elimination and inflammation or suppress inflammation and promote tissue remodeling and injury repair. During an infection, the clearance of dead and dying cells, a process named efferocytosis, can modulate the interplay between these contrasting functions. Here, we show that engulfment of SARS-CoV-2-infected apoptotic cells exacerbates inflammatory cytokine production, inhibits the expression of efferocytic receptors, and impairs continual efferocytosis by macrophages. We also provide evidence supporting that lung monocytes and macrophages from severe COVID-19 patients have compromised efferocytic capacity. Our findings reveal that dysfunctional efferocytosis of SARS-CoV-2-infected cell corpses suppresses macrophage anti-inflammation and efficient tissue repair programs and provides mechanistic insights for the excessive production of pro-inflammatory cytokines and accumulation of tissue damage associated with COVID-19 immunopathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
15. Molecular basis of carrageenan-induced cytokines production in macrophages
- Author
-
Alexandre H. Lopes, Rangel L. Silva, Miriam D. Fonseca, Francisco I. Gomes, Alexandre G. Maganin, Lucas S. Ribeiro, Lucas Maciel Mauriz Marques, Fernando Q. Cunha, Jose C. Alves-Filho, Dario S. Zamboni, Norberto P. Lopes, Bernardo S. Franklin, Aurélie Gombault, Fernando Silva Ramalho, Valerie F. J. Quesniaux, Isabelle Couillin, Bernhard Ryffel, and Thiago M. Cunha
- Subjects
Carrageenan ,Macrophages ,IL-1β ,NLRP3 Inflammasome ,Pannexin-1 channel ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Low molecular weight carrageenan (Cg) is a seaweed-derived sulfated polysaccharide widely used as inflammatory stimulus in preclinical studies. However, the molecular mechanisms of Cg-induced inflammation are not fully elucidated. The present study aimed to investigate the molecular basis involved in Cg-induced macrophages activation and cytokines production. Methods Primary culture of mouse peritoneal macrophages were stimulated with Kappa Cg. The supernatant and cell lysate were used for ELISA, western blotting, immunofluorescence. Cg-induced mouse colitis was also developed. Results Here we show that Cg activates peritoneal macrophages to produce pro-inflammatory cytokines such as TNF and IL-1β. While Cg-induced TNF production/secretion depends on TLR4/MyD88 signaling, the production of pro-IL-1β relies on TLR4/TRIF/SYK/reactive oxygen species (ROS) signaling pathway. The maturation of pro-IL1β into IL-1β is dependent on canonical NLRP3 inflammasome activation via Pannexin-1/P2X7/K+ efflux signaling. In vivo, Cg-induced colitis was reduced in mice in the absence of NLRP3 inflammasome components. Conclusions In conclusion, we unravel a critical role of the NLRP3 inflammasome in Cg-induced pro-inflammatory cytokines production and colitis, which is an important discovery on the pro-inflammatory properties of this sulfated polysaccharide for pre-clinical studies. Video abstract Graphical Abstract Carrageenan (Cg) is one the most used flogistic stimulus in preclinical studies. Nevertheless, the molecular basis of Cg-induced inflammation is not totally elucidated. Herein, Lopes et al. unraveled the molecular basis for Cg-induced macrophages production of biological active IL-1β. The Cg-stimulated macrophages produces pro-IL-1β depends on TLR4/TRIF/Syk/ROS, whereas its processing into mature IL-1β is dependent on the canonical NLRP3 inflammasome.
- Published
- 2020
- Full Text
- View/download PDF
16. Role of the transcriptional regulator SP140 in resistance to bacterial infections via repression of type I interferons
- Author
-
Daisy X Ji, Kristen C Witt, Dmitri I Kotov, Shally R Margolis, Alexander Louie, Victoria Chevée, Katherine J Chen, Moritz M Gaidt, Harmandeep S Dhaliwal, Angus Y Lee, Stephen L Nishimura, Dario S Zamboni, Igor Kramnik, Daniel A Portnoy, K Heran Darwin, and Russell E Vance
- Subjects
mycobacterium tuberculosis ,legionella pneumophila ,type i interferon ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Type I interferons (IFNs) are essential for anti-viral immunity, but often impair protective immune responses during bacterial infections. An important question is how type I IFNs are strongly induced during viral infections, and yet are appropriately restrained during bacterial infections. The Super susceptibility to tuberculosis 1 (Sst1) locus in mice confers resistance to diverse bacterial infections. Here we provide evidence that Sp140 is a gene encoded within the Sst1 locus that represses type I IFN transcription during bacterial infections. We generated Sp140–/– mice and found that they are susceptible to infection by Legionella pneumophila and Mycobacterium tuberculosis. Susceptibility of Sp140–/– mice to bacterial infection was rescued by crosses to mice lacking the type I IFN receptor (Ifnar–/–). Our results implicate Sp140 as an important negative regulator of type I IFNs that is essential for resistance to bacterial infections.
- Published
- 2021
- Full Text
- View/download PDF
17. Colchicine reduces the activation of NLRP3 inflammasome in COVID-19 patients
- Author
-
N. B. Amaral, T. S. Rodrigues, M. C. Giannini, M. I. Lopes, L. P. Bonjorno, P. I. S. O. Menezes, S. M. Dib, S. L. G. Gigante, M. N. Benatti, U. C. Rezek, L. L. Emrich-Filho, B. A. Sousa, S. C. L. Almeida, R. Luppino-Assad, F. P. Veras, A. H. Schneider, L. O. S. Leiria, L. D. Cunha, J. C. Alves-Filho, T. M. Cunha, E. Arruda, C. H. Miranda, A. Pazin-Filho, M. Auxiliadora-Martins, M. C. Borges, B. A. L. Fonseca, V. R. Bollela, C. M. Del-Ben, F. Q. Cunha, R. C. Santana, F. C. Vilar, D. S. Zamboni, P. Louzada-Junior, and R. D. R. Oliveira
- Subjects
Pharmacology ,Immunology - Published
- 2023
18. Interplay Between Reactive Oxygen Species and the Inflammasome Are Crucial for Restriction of Neospora caninum Replication
- Author
-
Caroline M. Mota, Djalma de S. Lima-Junior, Flávia Batista Ferreira França, Jhoan David Aguillón Torres, Patrício da Silva Cardoso Barros, Fernanda Maria Santiago, Joāo Santana Silva, José Roberto Mineo, Dario S. Zamboni, and Tiago W. P. Mineo
- Subjects
N. caninum ,ROS ,inflammasome ,macrophages ,mice ,Microbiology ,QR1-502 - Abstract
Neospora caninum poses as a considerable threat to animal health and generates significant economic impact in livestock production worldwide. Here, we have investigated the mechanism that underlies the participation of the inflammasome complex and Reactive Oxygen Species (ROS) in the regulation of immune responses during N. caninum infection. For that purpose, we used in vitro (bone marrow derived macrophages) and in vivo mouse models of infection. Our results show that NLRP3 and NLRC4 receptors, alongside with ASC and Caspase-1, are required for proper activation of the inflammasome during N. caninum infection. As expected, the engagement of these pathways is crucial for IL-1α, IL-1β, and IL-18 production, as well as the induction of pyroptosis. Our results also show that N. caninum induces ROS production dependent of the inflammasome assembly, which in its turn also depends on MyD88/NF-κB-induced ROS to maintain its activation and, ultimately, lead to restriction of parasite replication.
- Published
- 2020
- Full Text
- View/download PDF
19. Mitochondrial DNA Promotes NLRP3 Inflammasome Activation and Contributes to Endothelial Dysfunction and Inflammation in Type 1 Diabetes
- Author
-
Camila A. Pereira, Daniela Carlos, Nathanne S. Ferreira, Josiane F. Silva, Camila Z. Zanotto, Dario S. Zamboni, Valéria D. Garcia, Dora Fix Ventura, João S. Silva, and Rita C. Tostes
- Subjects
type 1 diabetes ,endothelial dysfunction ,NLRP3 inflammasome ,mitochondrial DNA ,inflammation ,reactive oxygen species ,Physiology ,QP1-981 - Abstract
Background: NLRP3 inflammasome activation in response to several signals, including mitochondrial DNA (mDNA), regulates inflammatory responses by caspase-1 activation and interleukin-1β (IL-1β) release. Circulating mDNA is linked to micro and macrovascular complications in diabetes. However, a role for mDNA in endothelial dysfunction is not clear. We tested the hypothesis that mDNA contributes to diabetes-associated endothelial dysfunction and vascular inflammation via NLRP3 activation.Methods: Vascular reactivity, reactive oxygen species (ROS) generation, calcium (Ca2+) influx and caspase-1 and IL-1β activation were determined in mesenteric resistance arteries from normoglicemic and streptozotocin-induced diabetic C57BL/6 and NLRP3 knockout (Nlrp3–/–) mice. Endothelial cells and mesenteric arteries were stimulated with mDNA from control (cmDNA) and diabetic (dmDNA) mice.Results: Diabetes reduced endothelium-dependent vasodilation and increased vascular ROS generation and caspase-1 and IL-1β activation in C57BL/6, but not in Nlrp3–/– mice. Diabetes increased pancreatic cytosolic mDNA. dmDNA decreased endothelium-dependent vasodilation. In endothelial cells, dmDNA activated NLRP3 via mitochondrial ROS and Ca2+ influx. Patients with type 1 diabetes exhibited increased circulating mDNA as well as caspase-1 and IL-1β activation.Conclusion: dmDNA activates endothelial NLRP3 inflammasome by mechanisms that involve Ca2+ influx and mitochondrial ROS generation. NLRP3 deficiency prevents diabetes-associated vascular inflammatory damage and endothelial dysfunction. Our study highlights the importance of NLRP3 inflammasome in diabetes-associated vascular dysfunction, which is key to diabetic complications.
- Published
- 2020
- Full Text
- View/download PDF
20. AIM2 Engages Active but Unprocessed Caspase-1 to Induce Noncanonical Activation of the NLRP3 Inflammasome
- Author
-
Larissa D. Cunha, Alexandre L.N. Silva, Juliana M. Ribeiro, Danielle P.A. Mascarenhas, Gustavo F.S. Quirino, Leonardo L. Santos, Richard A. Flavell, and Dario S. Zamboni
- Subjects
AIM2 ,caspase-1 ,inflammasomes ,Legionella pneumophila ,NLRP3 ,Biology (General) ,QH301-705.5 - Abstract
Inflammasomes are multimeric protein complexes that initiate inflammatory cascades. Their activation is a hallmark of many infectious or inflammatory diseases. Their composition and activity are specified by proinflammatory stimuli. For example, the NLRP3 inflammasome is activated in response to cell damage and K+ efflux, whereas the AIM2 inflammasome is activated in response to cytosolic DNA. We used Legionella pneumophila, an intracellular bacterial pathogen that activates multiple inflammasomes, to elucidate the molecular mechanisms regulating inflammasome activation during infection. Upon infection, the AIM2 inflammasome engaged caspase-1 to induce pore formation in the cell membrane, which then caused K+-efflux-mediated activation of NLRP3. Thus, the AIM2 inflammasome amplifies signals of infection, triggering noncanonical activation of NLRP3. During infection, AIM2 and caspase-11 induced membrane damage, which was sufficient and essential for activating the NLRP3 inflammasome. Our data reveal that different inflammasomes regulate one another’s activity to ensure an effective immune response to infection.
- Published
- 2017
- Full Text
- View/download PDF
21. Leishmania Lipophosphoglycan Triggers Caspase-11 and the Non-canonical Activation of the NLRP3 Inflammasome
- Author
-
Renan V.H. de Carvalho, Warrison A. Andrade, Djalma S. Lima-Junior, Marisa Dilucca, Caroline V. de Oliveira, Kun Wang, Paula M. Nogueira, Jeronimo N. Rugani, Rodrigo P. Soares, Stephen M. Beverley, Feng Shao, and Dario S. Zamboni
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Activation of the NLRP3 inflammasome by Leishmania parasites is critical for the outcome of leishmaniasis, a disease that affects millions of people worldwide. We investigate the mechanisms involved in NLRP3 activation and demonstrate that caspase-11 (CASP11) is activated in response to infection by Leishmania species and triggers the non-canonical activation of NLRP3. This process accounts for host resistance to infection in macrophages and in vivo. We identify the parasite membrane glycoconjugate lipophosphoglycan (LPG) as the molecule involved in CASP11 activation. Cytosolic delivery of LPG in macrophages triggers CASP11 activation, and infections performed with Lpg1–/– parasites reduce CASP11/NLRP3 activation. Unlike bacterial LPS, purified LPG does not activate mouse CASP11 (or human Casp4) in vitro, suggesting the participation of additional molecules for LPG-mediated CASP11 activation. Our data identify a parasite molecule involved in CASP11 activation, thereby establishing the mechanisms underlying inflammasome activation in response to Leishmania species. : Activation of the NLRP3 inflammasome is critical for the outcome of leishmaniasis. De Carvalho et al. show that Leishmania lipophosphoglycan (LPG) triggers caspase-11 activation in macrophage cytoplasm and the non-canonical activation of NLRP3, thereby establishing the mechanisms underlying NLRP3 activation in response to Leishmania. Keywords: Leishmania, inflammasome, caspase-11, lipophosphoglycan, LPG, caspase-1, NLRP3, macrophage, non-canonical inflammasome, Leishmaniasis
- Published
- 2019
- Full Text
- View/download PDF
22. TONSILS ARE MAJOR SITES OF PROLONGED SARS-COV-2 INFECTION IN CHILDREN
- Author
-
Thais M. Lima, Ronaldo B. Martins, Carolina S. Miura, Maria V. O. Souza, Murilo H. A. Cassiano, Tamara S. Rodrigues, Flávio P. Veras, Josane F. Sousa, Rogério Gomes, Glaucia M. Almeida, Stella R. Melo, Gabriela C. Silva, Matheus Dias, Carlos F. Capato, Maria L. Silva, Veridiana E. D. Barros, Lucas R. Carenzi, Dario S. Zamboni, Daniel M. M. Jorge, Edwin Tamashiro, Wilma T. Anselmo-Lima, Fabiana C. P. Valera, and Eurico Arruda
- Abstract
In the present study, we show that SARS-CoV-2 can infect palatine tonsils and adenoids in children without symptoms of COVID-19, with no history of recent upper airway infection. We studied 48 children undergoing tonsillectomy due to snoring/OSA or recurrent tonsillitis between October 2020 and September 2021. Briefly, nasal cytobrush (NC), nasal wash (NW) and tonsillar tissue fragments obtained at surgery were tested by RT-PCR, immunohistochemistry (IHC), flow cytometry and neutralization assay. We detected the presence of SARS-CoV-2 in at least one specimen tested in 25% of patients (20% in palatine tonsils and 16.27% in adenoids, 10.41% of NC and 6.25% of NW). Importantly, in 2 of the children there was evidence of laboratory-confirmed acute infection 2 and 5 months before surgery. IHC revealed the presence of SARS-CoV-2 nucleoprotein in epithelial surface and in lymphoid cells in both extrafollicular and follicular regions, in adenoids and palatine tonsils. Flow cytometry showed that CD20+B lymphocytes were the most infected phenotypes by SARS-CoV-2 NP, followed by CD4+ and CD8+ T lymphocytes, and CD14+ macrophages and dendritic cells. Additionally, IF indicated that SARS-CoV-2-infected tonsillar tissues had increased expression of ACE2 and TMPRSS2. NGS sequencing demonstrated the presence of different SARS CoV-2 variants in tonsils from different tissues. SARS-CoV-2 antigen detection was not restricted to tonsils, but was also detected in nasal cells from the olfactory region. In conclusion, palatine tonsils and adenoids are sites of prolonged infection by SARS-CoV-2 in children, even without COVID-19 symptoms.
- Published
- 2023
23. Editor's evaluation: Formation and three-dimensional architecture of Leishmania adhesion in the sand fly vector
- Author
-
Dario S Zamboni
- Published
- 2023
24. Caspase-8 but not caspase-7 influences inflammasome activation to act in control of Brucella abortus infection
- Author
-
Raiany A. Santos, Daiane M. Cerqueira, Dario S. Zamboni, and Sergio C. Oliveira
- Subjects
Microbiology (medical) ,Microbiology - Abstract
Programmed cell death (PCD) is an important mechanism of innate immunity against bacterial pathogens. The innate immune PCD pathway involves the molecules caspase-7 and caspase-8, among others. Brucella abortus is a gram-negative bacterium that causes a zoonotic disease termed brucellosis. The innate immune response against this pathogen involves activation of inflammasome components and induction of pyroptosis. However, no studies so far have revealed the role of caspase-7 or caspase-8 during this bacterial infection. Herein, we demonstrate that caspase-7 is dispensable for caspase-1 processing, IL-1β secretion and cell death in macrophages. Additionally, caspase-7 deficient animals control B. abortus infection as well as the wild type mice. Furthermore, we addressed the role of caspase-8 in inflammasome activation and pyroptosis during this bacterial infection. Macrophages deficient in caspase-8 secreted reduced amounts of IL-1β that parallels with diminished caspase-1 activity when compared to wild type cells. Additionally, caspase-8 KO macrophages showed reduced LDH release when compared to wild type, suggesting that caspase-8 may play an important role in pyroptosis in response to B. abortus. Finally, caspase-8 KO animals were more susceptible to Brucella infection when compared to wild type mice. Overall, this study contributes to a better understanding of the involvement of caspase-7 and caspase-8 in innate immunity against B. abortus infection.
- Published
- 2022
25. Cell death induced by NLRP3-palmitate axis impairs pulmonary damage tolerance and aggravates immunopathology during obesity-tuberculosis comorbidity
- Author
-
Sandra P Palma Albornoz, Thais FC Fraga‐Silva, Renan VH de Carvalho, Tamara S Rodrigues, Ana Flávia Gembre, Rômulo Silva de Oliveira, Fernanda Mesquita de Souza, Giseli Furlan Corrêa, Leandra NZ Ramalho, Daniela Carlos, Danilo C de Almeida, Niels OS Câmara, Dario S Zamboni, Viviani Nardini Takahashi, Carlos A Sorgi, Lucia H Faccioli, Alexandra I Medeiros, Diego Luís Costa, and Vânia LD Bonato
- Subjects
IMUNOLOGIA ,Pathology and Forensic Medicine - Abstract
A low grade and persistent inflammation, which is the hallmark of obesity, requires the participation of NLRP3 and cell death. During Mycobacterium tuberculosis infection, NLRP3 signaling is important for bacterial killing by macrophages in vitro, but was shown to be dispensable for host protection in vivo. We hypothesized that during obesity-tuberculosis comorbidity, NLRP3 signaling might play a detrimental role by inducing excessive inflammation. We employed a model of high fat diet-induced obesity, followed by M. tuberculosis infection in C57BL/6 mice. Obese mice presented increased susceptibility to infection and pulmonary immunopathology compared to lean mice. Using treatment with NLRP3 antagonist and Nlrp3
- Published
- 2022
26. Author response for 'Inflammasome activation and CCR2‐mediated monocyte‐derived dendritic cell recruitment restrict Legionella pneumophila infection'
- Author
-
null Marco A. Ataide, null Graziele Z. Manin, null Samuel S. Oliveira, null O. RhanoicaGuerra, and null Dario S. Zamboni
- Published
- 2022
27. Identification of immunomodulatory drugs that inhibit multiple inflammasomes and impair SARS-CoV-2 infection
- Author
-
Letícia de Almeida, Alexandre L. N. da Silva, Tamara S. Rodrigues, Samuel Oliveira, Adriene Y. Ishimoto, Amanda A. Seribelli, Amanda Becerra, Warrison A. Andrade, Marco A. Ataide, Camila C. S. Caetano, Keyla S. G. de Sá, Natália Pelisson, Ronaldo B. Martins, Juliano de Paula Souza, Eurico Arruda, Sabrina S. Batah, Ricardo Castro, Fabiani G. Frantz, Fernando Q. Cunha, Thiago M. Cunha, Alexandre T. Fabro, Larissa D. Cunha, Paulo Louzada-Junior, Rene D. R. de Oliveira, and Dario S. Zamboni
- Subjects
Immunomodulating Agents ,Mice ,Multidisciplinary ,Inflammasomes ,SARS-CoV-2 ,NLR Family, Pyrin Domain-Containing 3 Protein ,Animals ,Humans ,COVID-19 Drug Treatment - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces mild or asymptomatic COVID-19 in most cases, but some patients develop an excessive inflammatory process that can be fatal. As the NLRP3 inflammasome and additional inflammasomes are implicated in disease aggravation, drug repositioning to target inflammasomes emerges as a strategy to treat COVID-19. Here, we performed a high-throughput screening using a 2560 small-molecule compound library and identified FDA-approved drugs that function as pan-inflammasome inhibitors. Our best hit, niclosamide (NIC), effectively inhibits both inflammasome activation and SARS-CoV-2 replication. Mechanistically, induction of autophagy by NIC partially accounts for inhibition of NLRP3 and AIM2 inflammasomes, but NIC-mediated inhibition of NAIP/NLRC4 inflammasome are autophagy independent. NIC potently inhibited inflammasome activation in human monocytes infected in vitro, in PBMCs from patients with COVID-19, and in vivo in a mouse model of SARS-CoV-2 infection. This study provides relevant information regarding the immunomodulatory functions of this promising drug for COVID-19 treatment.
- Published
- 2022
28. Inhibition of caspase-1 or gasdermin-D enable caspase-8 activation in the Naip5/NLRC4/ASC inflammasome.
- Author
-
Danielle P A Mascarenhas, Daiane M Cerqueira, Marcelo S F Pereira, Fernanda V S Castanheira, Talita D Fernandes, Graziele Z Manin, Larissa D Cunha, and Dario S Zamboni
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Legionella pneumophila is a Gram-negative, flagellated bacterium that survives in phagocytes and causes Legionnaires' disease. Upon infection of mammalian macrophages, cytosolic flagellin triggers the activation of Naip/NLRC4 inflammasome, which culminates in pyroptosis and restriction of bacterial replication. Although NLRC4 and caspase-1 participate in the same inflammasome, Nlrc4-/- mice and their macrophages are more permissive to L. pneumophila replication compared with Casp1/11-/-. This feature supports the existence of a pathway that is NLRC4-dependent and caspase-1/11-independent. Here, we demonstrate that caspase-8 is recruited to the Naip5/NLRC4/ASC inflammasome in response to flagellin-positive bacteria. Accordingly, caspase-8 is activated in Casp1/11-/- macrophages in a process dependent on flagellin, Naip5, NLRC4 and ASC. Silencing caspase-8 in Casp1/11-/- cells culminated in macrophages that were as susceptible as Nlrc4-/- for the restriction of L. pneumophila replication. Accordingly, macrophages and mice deficient in Asc/Casp1/11-/- were more susceptible than Casp1/11-/- and as susceptible as Nlrc4-/- for the restriction of infection. Mechanistically, we found that caspase-8 activation triggers gasdermin-D-independent pore formation and cell death. Interestingly, caspase-8 is recruited to the Naip5/NLRC4/ASC inflammasome in wild-type macrophages, but it is only activated when caspase-1 or gasdermin-D is inhibited. Our data suggest that caspase-8 activation in the Naip5/NLRC4/ASC inflammasome enable induction of cell death when caspase-1 or gasdermin-D is suppressed.
- Published
- 2017
- Full Text
- View/download PDF
29. Targeting C5aR1 signaling reduced neutrophil extracellular traps and ameliorates COVID-19 pathology
- Author
-
Bruna M. Silva, Flavio P. Veras, Giovanni F. Gomes, Seppe Cambier, Gabriel V. L. Silva, Andreza U. Quadros, Diego B. Caetité, Daniele C. Nascimento, Camilla M. Silva, Juliana C. Silva, Samara Damasceno, Ayda H. Schneider, Fabio Beretta, Sabrina S. Batah, Icaro M. S. Castro, Isadora M. Paiva, Tamara Rodrigues, Ana Salina, Ronaldo Martins, Guilherme C.M. Cebinelli, Naira L. Bibo, Daniel M. Jorge, Helder I. Nakaya, Dario S. Zamboni, Luiz O. Leiria, Alexandre T. Fabro, José C. Alves-Filho, Eurico Arruda, Paulo Louzada-Junior, Renê D. Oliveira, Larissa D. Cunha, Pierre Van Mol, Lore Vanderbeke, Simon Feys, Els Wauters, Laura Brandolini, Fernando Q. Cunha, Jörg Köhl, Marcello Allegretti, Diether Lambrechts, Joost Wauters, Paul Proost, and Thiago M. Cunha
- Abstract
Patients with severe COVID-19 develop acute respiratory distress syndrome (ARDS) that may progress to cytokine storm syndrome, organ dysfunction, and death. Considering that complement component 5a (C5a), through its cellular receptor C5aR1, has potent proinflammatory actions, and plays immunopathological roles in inflammatory diseases, we investigated whether C5a/C5aR1 pathway could be involved in COVID-19 pathophysiology. C5a/C5aR1 signaling increased locally in the lung, especially in neutrophils of critically ill COVID-19 patients compared to patients with influenza infection, as well as in the lung tissue of K18-hACE2 Tg mice (Tg mice) infected with SARS-CoV-2. Genetic and pharmacological inhibition of C5aR1 signaling ameliorated lung immunopathology in Tg-infected mice. Mechanistically, we found that C5aR1 signaling drives neutrophil extracellular trap (NET)s-dependent immunopathology. These data confirm the immunopathological role of C5a/C5aR1 signaling in COVID-19 and indicate that antagonist of C5aR1 could be useful for COVID-19 treatment.
- Published
- 2022
30. CASP4/11 contributes to pulmonary inflammation and disease exacerbation in COVID-19
- Author
-
Tamara S. Rodrigues, Camila C.S. Caetano, Keyla S.G. de Sá, Leticia Almeida, Amanda Becerra, Augusto V. Gonçalves, Leticia de Sousa Lopes, Samuel Oliveira, Danielle P.A. Mascarenhas, Sabrina S. Batah, Bruna M. Silva, Giovanni F. Gomes, Ricardo Castro, Ronaldo B. Martins, Jonathan Avila, Fabiani G. Frantz, Thiago M. Cunha, Eurico Arruda, Fernando Q Cunha, Helder Nakaya, Larissa D. Cunha, Alexandre T Fabro, Paulo Louzada-Junior, Renê D.R. de Oliveira, and Dario S. Zamboni
- Abstract
Infection with SARS-CoV-2 induces COVID-19, an inflammatory disease that is usually self-limited, but depending on patient conditions may culminate with critical illness and patient death. The virus triggers activation of intracellular sensors, such as the NLRP3 inflammasome, which promotes inflammation and aggravates the disease. Thus, identification of host components associated with NLRP3 inflammasome is key for understanding the physiopathology of the disease. Here, we reported that SARS-CoV-2 induces upregulation and activation of human Caspase-4/CASP4 (mouse Caspase-11/CASP11) and this process contributes to inflammasome activation in response to SARS-CoV-2. CASP4 was expressed in lung autopsy of lethal cases of COVID-19 and CASP4 expression correlates with expression of inflammasome components and inflammatory mediators such as CASP1, IL1B, IL18 and IL6. In vivo infections performed in transgenic hACE2 humanized mouse, deficient or sufficient for Casp11, indicate that hACE2 Casp11−/− mice were protected from disease development, with reduced body weight loss, reduced temperature variation, increased pulmonary parenchymal area, reduced clinical score of the disease and reduced mortality. Collectively, our data establishes that CASP4/11 contributes to disease pathology and contributes for future immunomodulatory therapeutic interventions to COVID-19.
- Published
- 2022
31. Inflammasome activation and pulmonary viral loads define two distinct clinical outcomes in COVID-19
- Author
-
Keyla S.G. de Sá, Luana A. Amaral, Camila C.S. Caetano, Amanda Becerra, Sabrina S. Batah, Isadora M. de Oliveira, Letícia S. Lopes, Leticia Almeida, Samuel Oliveira, Danilo Tadao Wada, Marcel Koenigkam-Santos, Ronaldo B. Martins, Roberta R. C. Rosales, Eurico Arruda, Alexandre T Fabro, and Dario S. Zamboni
- Abstract
COVID-19 has affected more than half a billion people worldwide, with more than 6.3 million deaths, but the pathophysiological mechanisms involved in lethal cases and the host determinants that determine the different clinical outcomes are still unclear. In this study, we assessed lung autopsies of 47 COVID-19 patients and examined the inflammatory profiles, viral loads, and inflammasome activation. Additionally, we correlated these factors with the patient’s clinical and histopathological conditions. Robust inflammasome activation, mediated by macrophages and endothelial cells, was detected in the lungs of lethal cases of SARS-CoV-2. An analysis of gene expression allowed for the classification of COVID-19 patients into two different clusters. Cluster 1 died with higher viral loads and exhibited a reduced inflammatory profile than Cluster 2. Illness time, mechanical ventilation time, pulmonary fibrosis, respiratory functions, histopathological status, thrombosis, viral loads and inflammasome activation significantly differed between the two clusters. Our data demonstrated two distinct profiles in lethal cases of COVID-19, thus indicating that the balance of viral replication and inflammasome-mediated pulmonary inflammation led to different clinical outcomes. We provide important information to understand clinical variations in severe COVID-19, a process that is critical for decisions between immune-mediated or antiviral-mediated therapies for the treatment of critical cases of COVID-19.
- Published
- 2022
32. Dietary Fiber Drives IL-1β–Dependent Peritonitis Induced by Bacteroides fragilis via Activation of the NLRP3 Inflammasome
- Author
-
Juliana Echevarria-Lima, Leandro Araujo Lobo, Charles R. Mackay, Dario S. Zamboni, Robson Coutinho-Silva, Ana Carolina Oliveira, Julio Scharfstein, Regina Maria Cavalcanti Pilotto Domingues, Aline Cristina Abreu Moreira-Souza, Eliana Mariño, Erivan Schnaider Ramos-Junior, Maria Bellio, Eliane de Oliveira Ferreira, Juliana P Castelpoggi, and Bruno Jennings-Almeida
- Subjects
biology ,Chemistry ,Immunology ,Peritonitis ,Inflammasome ,Context (language use) ,biology.organism_classification ,medicine.disease ,Microbiology ,Proinflammatory cytokine ,03 medical and health sciences ,Peritoneal cavity ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,medicine ,Immunology and Allergy ,Secretion ,Bacteroides fragilis ,030215 immunology ,medicine.drug - Abstract
Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1β contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1β secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1β–dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1β secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1β production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1β production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1β–dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R–deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1β secretion outside the gut.
- Published
- 2021
33. Author response: Efferocytosis of SARS-CoV-2-infected dying cells impairs macrophage anti-inflammatory functions and clearance of apoptotic cells
- Author
-
Tamara S Rodrigues, Douglas dos-Santos, Ana CG Salina, Marlon Fortes-Rocha, Edismauro G Freitas-Filho, Daniel L Alzamora-Terrel, Icaro MS Castro, Thais FC Fraga da Silva, Mikhael HF de Lima, Daniele C Nascimento, Camila M Silva, Juliana E Toller-Kawahisa, Amanda Becerra, Samuel Oliveira, Diego B Caetité, Leticia Almeida, Adriene Y Ishimoto, Thais M Lima, Ronaldo B Martins, Flavio Veras, Natália B do Amaral, Marcela C Giannini, Letícia P Bonjorno, Maria IF Lopes, Maira N Benatti, Sabrina S Batah, Rodrigo C Santana, Fernando C Vilar, Maria A Martins, Rodrigo L Assad, Sergio CL de Almeida, Fabiola R de Oliveira, Eurico Arruda Neto, Thiago M Cunha, José C Alves-Filho, Vania LD Bonato, Fernando Q Cunha, Alexandre T Fabro, Helder I Nakaya, Dario S Zamboni, Paulo Louzada-Junior, Rene DR Oliveira, and Larissa D Cunha
- Published
- 2022
34. SARS-CoV-2 productively infects primary human immune system cells in vitro and in COVID-19 patients
- Author
-
Marjorie C Pontelli, Ítalo A Castro, Ronaldo B Martins, Leonardo La Serra, Flávio P Veras, Daniele C Nascimento, Camila M Silva, Ricardo S Cardoso, Roberta Rosales, Rogério Gomes, Thais M Lima, Juliano P Souza, Brenda C Vitti, Diego B Caetité, Mikhael H F de Lima, Spencer D Stumpf, Cassandra E Thompson, Louis-Marie Bloyet, Juliana E Toller-Kawahisa, Marcela C Giannini, Letícia P Bonjorno, Maria I F Lopes, Sabrina S Batah, Li Siyuan, Rodrigo Luppino-Assad, Sergio C L Almeida, Fabiola R Oliveira, Maíra N Benatti, Lorena L F Pontes, Rodrigo C Santana, Fernando C Vilar, Maria Auxiliadora-Martins, Pei-Yong Shi, Thiago M Cunha, Rodrigo T Calado, José C Alves-Filho, Dario S Zamboni, Alexandre T Fabro, Paulo Louzada-Junior, Rene D R Oliveira, Sean P J Whelan, Fernando Q Cunha, and Eurico Arruda
- Subjects
viruses ,Genetics ,Cell Biology ,General Medicine ,Molecular Biology - Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with a hyperinflammatory state and lymphocytopenia, a hallmark that appears as both signature and prognosis of disease severity outcome. Although cytokine storm and a sustained inflammatory state are commonly associated with immune cell depletion, it is still unclear whether direct SARS-CoV-2 infection of immune cells could also play a role in this scenario by harboring viral replication. We found that monocytes, as well as both B and T lymphocytes, were susceptible to SARS-CoV-2 infection in vitro, accumulating double-stranded RNA consistent with viral RNA replication and ultimately leading to expressive T cell apoptosis. In addition, flow cytometry and immunofluorescence analysis revealed that SARS-CoV-2 was frequently detected in monocytes and B lymphocytes from coronavirus disease 2019 (COVID-19) patients. The rates of SARS-CoV-2-infected monocytes in peripheral blood mononuclear cells from COVID-19 patients increased over time from symptom onset, with SARS-CoV-2-positive monocytes, B cells, and CD4+ T lymphocytes also detected in postmortem lung tissue. These results indicated that SARS-CoV-2 infection of blood-circulating leukocytes in COVID-19 patients might have important implications for disease pathogenesis and progression, immune dysfunction, and virus spread within the host.
- Published
- 2022
35. Leishmania Viannia guyanensis, LRV1 virus and extracellular vesicles: a dangerous trio influencing the faith of immune response during muco-cutaneous leishmaniasis
- Author
-
Dario S. Zamboni and Martin Olivier
- Subjects
Leishmaniasis, Mucocutaneous ,0301 basic medicine ,Leishmaniavirus ,Immunology ,Virulence ,Virus ,Extracellular Vesicles ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cutaneous leishmaniasis ,parasitic diseases ,medicine ,Animals ,Humans ,Immunology and Allergy ,Macrophage ,Leishmania guyanensis ,biology ,Leishmaniasis ,RNA virus ,medicine.disease ,Leishmania ,biology.organism_classification ,Virology ,3. Good health ,030104 developmental biology ,RNA ,030215 immunology - Abstract
Parasites of Leishmania genus have developed various strategies to overcome host immune response favoring its infection and development toward leishmaniasis. With an array of virulence factors, those parasites modify host macrophage signaling and functions. Depending of the species involved, visceral or cutaneous leishmaniasis will develop. Several years ago, Leishmania Viannia guyanensis that is naturally infected with the endosymbiotic virus Leishmania RNA Virus 1 was found to cause a particularly aggressive form of South-American mucocutaneous leishmaniasis. This virus, when co-transmitted with the parasite was shown to strongly modulate RNA sensors and NLRP3 inflammasome network that could explain in part the exacerbated skin pathology caused by this particular parasite. In this review, we will be discussing how this endosymbiotic virus-infected Leishmania in conjunction with Leishmania exosomes partner together to manipulate host immune response in their favor.
- Published
- 2020
36. Caspase-8 mediates inflammation and disease in rodent malaria
- Author
-
Larissa M. N. Pereira, Katherine A. Fitzgerald, Caroline Junqueira, Patricia A. Assis, Ricardo T. Gazzinelli, Dario S. Zamboni, Dhelio Batista Pereira, Natalia M. de Araujo, Danielle Fernandes Durso, Egil Lien, Douglas T. Golenbock, and Marco Antonio Ataide
- Subjects
0301 basic medicine ,Science ,General Physics and Astronomy ,Inflammation ,Systemic inflammation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,parasitic diseases ,medicine ,MALÁRIA ,lcsh:Science ,Caspase ,Multidisciplinary ,biology ,business.industry ,Inflammasome ,General Chemistry ,medicine.disease ,Malaria ,030104 developmental biology ,Cerebral Malaria ,Immunology ,biology.protein ,Infectious diseases ,Tumor necrosis factor alpha ,lcsh:Q ,medicine.symptom ,business ,Parasite host response ,030215 immunology ,medicine.drug - Abstract
Earlier studies indicate that either the canonical or non-canonical pathways of inflammasome activation have a limited role on malaria pathogenesis. Here, we report that caspase-8 is a central mediator of systemic inflammation, septic shock in the Plasmodium chabaudi-infected mice and the P. berghei-induced experimental cerebral malaria (ECM). Importantly, our results indicate that the combined deficiencies of caspases-8/1/11 or caspase-8/gasdermin-D (GSDM-D) renders mice impaired to produce both TNFα and IL-1β and highly resistant to lethality in these models, disclosing a complementary, but independent role of caspase-8 and caspases-1/11/GSDM-D in the pathogenesis of malaria. Further, we find that monocytes from malaria patients express active caspases-1, -4 and -8 suggesting that these inflammatory caspases may also play a role in the pathogenesis of human disease., Inflammasome activation plays a role in malaria pathogenesis, but details aren’t well understood. Here, the authors show that caspase-8 is a central mediator of systemic inflammation in rodent malaria and that monocytes from malaria patients express active caspases-1, -4 and -8.
- Published
- 2020
37. NLRP12 controls arthritis severity by acting as a checkpoint inhibitor of Th17 cell differentiation
- Author
-
Fernando Q. Cunha, Douglas Silva Prado, Flávio P. Veras, Thiago M. Cunha, Dario S. Zamboni, Luis Eduardo Alves Damasceno, Paulo Henrique Melo, José C. Alves-Filho, and Raphael Gomes Ferreira
- Subjects
Male ,STAT3 Transcription Factor ,0301 basic medicine ,Adoptive cell transfer ,FATORES DE TRANSCRIÇÃO ,T cell ,Cellular differentiation ,Cell ,Population ,Arthritis ,Inflammation ,Nod ,Biochemistry ,Arthritis, Rheumatoid ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,education ,Molecular Biology ,education.field_of_study ,business.industry ,Interleukin-17 ,Intracellular Signaling Peptides and Proteins ,Cell Differentiation ,medicine.disease ,Arthritis, Experimental ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Neutrophil Infiltration ,Cancer research ,Th17 Cells ,Joints ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Nucleotide oligomerization domain (NOD)-like receptor-12 (NLRP12) has emerged as a negative regulator of inflammation. It is well described that the Th17 cell population increases in patients with early Rheumatoid Arthritis (RA), which correlates with the disease activity. Here, we investigated the role of NLRP12 in the differentiation of Th17 cells and the development of experimental arthritis, using the antigen-induced arthritis (AIA) murine model. We found that Nlrp12-/ - mice develop severe arthritis characterized by an exacerbated Th17-mediated inflammatory response with increases in the articular hyperalgesia, knee joint swelling, and neutrophil infiltration. Adoptive transfer of Nlrp12-/ - cells into WT mice recapitulated the hyperinflammatory response seen in Nlrp12-/ - mice and the treatment with anti-IL-17A neutralizing antibody abrogated arthritis development in Nlrp12-/ - mice, suggesting that NLRP12 works as an inhibitor of Th17 cell differentiation. Indeed, Th17 cell differentiation markedly increases in Nlrp12-/- T cells cultured under the Th17-skewing condition. Mechanistically, we found that NLRP12 negatively regulates IL-6-induced phosphorylation of STAT3 in T cells. Finally, pharmacological inhibition of STAT3 reduced Th17 cell differentiation and abrogated hyperinflammatory arthritis observed in Nlrp12-/ - mice. Thus, we described a novel role for NLRP12 as a checkpoint inhibitor of Th17 cell differentiation, which controls the severity of experimental arthritis.
- Published
- 2020
38. NLRC4 biology in immunity and inflammation
- Author
-
Warrison A. Andrade and Dario S. Zamboni
- Subjects
0301 basic medicine ,Inflammasomes ,Immunology ,Caspase 1 ,Inflammation ,Biology ,Autoimmune Diseases ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,NLRC4 ,medicine ,Animals ,Humans ,Immunology and Allergy ,Caspase 7 ,Caspase 8 ,Innate immune system ,Effector ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,Pyroptosis ,Inflammasome ,Bacterial Infections ,Cell Biology ,Phosphate-Binding Proteins ,DOENÇAS AUTOIMUNES ,Cell biology ,CARD Signaling Adaptor Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,medicine.symptom ,medicine.drug - Abstract
Inflammasomes are cytosolic multiprotein complexes that sense microbial infections or host cell damage, triggering cytokine production and a proinflammatory form of cell death, called pyroptosis. Whereas pyroptosis and cytokine production may often promote host resistance to infections, uncontrolled inflammasome activation leads to autoinflammatory diseases in humans. Among the multiple inflammasomes described, the neuronal apoptosis inhibitory protein/nucleotide-binding domain leucine-rich repeat-containing protein family caspase activation and recruitment domain-containing protein 4 (NLRC4) inflammasome emerged as a critical component for the restriction of bacterial infections. Accordingly, our understanding of this inflammasome advanced remarkably over the last 10 yr, expanding our knowledge about ligand-receptor interaction; cryo-EM structure; and downstream effectors and substrates, such as gasdermin-D, caspase-1, caspase-8, and caspase-7. In this review, we discuss recent advances on the biology of the NLRC4 inflammasome, in terms of structure and activation mechanisms, importance in bacterial and nonbacterial diseases, and the identification of NLRC4 gain-of-function mutations leading to NLRC4-associated autoinflammatory diseases in humans.
- Published
- 2020
39. Gasdermin-D activation by SARS-CoV-2 trigger NET and mediate COVID-19 immunopathology
- Author
-
Camila Meirelles Silva, Carlos Wagner S Wanderley, Flavio Protasio Veras, Augusto Veloso Gonçalves, Mikhael Haruo Fernandes Lima, Juliana E. Toller Kawahisa, Giovanni Freitas Gomes, Daniele Carvalho Nascimento, Valter V. Silva Monteiro, Isadora Marques Paiva, Cícero José Luíz Ramos Almeida, Diego Brito Caetité, Juliana da Costa Silva, Maria Isabel Fernandes Lopes, Letícia Pastorelli Bonjorno, Marcela Cavichioli Giannini, Natalia Brasil Amaral, Maíra Nilson Benatti, Luis Eduardo Alves Damasceno, Bruna Manuella Souza Silva, Ayda Henriques Schneider, Icaro Maia Santos Castro, Juan Carlo Santos Silva, Amanda Pereira Vasconcelos, Tiago Tomazini Gonçalves, Sabrina Setembre Batah, Tamara Silva Rodrigues, Victor Ferreira Costa, Marjorie Cornejo Pontelli, Ronaldo B Martins, Timna Varela Martins, Danillo Lucas Alves Espósito, Guilherme Cesar Martelossi Cebinelli, Benedito Antônio Lopes da Fonseca, Luiz Osório Silveira Leiria, Larissa Dias Cunha, Eurico Arruda, Helder I Nakaia, Alexandre Todorovic Fabro, Renê D Oliveira, Dario S Zamboni, Paulo Louzada Junior, Thiago Mattar Cunha, José Carlos Farias Alves Filho, and Fernando de Queiroz Cunha
- Abstract
The release of neutrophil extracellular traps (NETs) is associated with inflammation, coagulopathy, and organ damage found in severe cases of COVID-19. However, the molecular mechanisms underlying the release of NETs in COVID-19 remain unclear. Using a single-cell transcriptome analysis we observed that the expression of GSDMD and inflammasome-related genes were increased in neutrophils from COVID-19 patients. Furthermore, high expression of GSDMD was found associated with NETs structures in the lung tissue of COVID-19 patients. The activation of GSDMD in neutrophils requires live SARS-CoV-2 and occurs after neutrophil infection via ACE2 receptors and serine protease TMPRSS2. In a mouse model of SARS-CoV-2 infection, the treatment with GSDMD inhibitor (disulfiram) reduced NETs release and organ damage. These results demonstrated that GSDMD-dependent NETosis plays a critical role in COVID-19 immunopathology, and suggests that GSDMD inhibitors, can be useful to COVID-19 treatment.In BriefHere, we showed that the activation of the Gasdermin-D (GSDMD) pathway in neutrophils controls NET release during COVID-19. The inhibition of GSDMD with disulfiram, abrogated NET formation reducing lung inflammation and tissue damage. These findings suggest GSDMD as a target for improving the COVID-19 therapy.
- Published
- 2022
40. Sepsis-induced immunosuppression is marked by an expansion of a highly suppressive repertoire of FOXP3+ T-regulatory cells expressing TIGIT
- Author
-
Fernando Q. Cunha, Carlos W. S. Wanderley, Carlos Hiroji Hiroki, Camilla Meireles S Silva, Augusto V. Gonçalves, Gustavo Fernando da Silva Quirino, Guilherme Cesar Martelossi Cebinelli, Marcos Henrique Rosa, Thiago M. Cunha, Dario S. Zamboni, Jessica Adrielle Teixeira Santos, José-Carlos A Filho, Mikhael Haruo Fernandes de Lima, and Vanessa F. Borges
- Subjects
medicine.medical_treatment ,Biology ,T-Lymphocytes, Regulatory ,Flow cytometry ,Sepsis ,Mice ,TIGIT ,medicine ,Animals ,Immunology and Allergy ,Receptors, Immunologic ,STAT6 ,Immunosuppression Therapy ,TNFRSF18 ,medicine.diagnostic_test ,FOXP3 ,Forkhead Transcription Factors ,Immunosuppression ,Interleukin-33 ,medicine.disease ,Interleukin-1 Receptor-Like 1 Protein ,Interleukin 33 ,CITOMETRIA DE FLUXO ,Infectious Diseases ,Immunology - Abstract
Background Although the literature shows that an increase in both the number and suppressive function of CD4+forkhead box P3 (FOXP3)+ T-regulatory cells (Tregs) during sepsis contributes to an immunosuppressed state, little is known about the identity of these cells. Methods Using the sepsis mouse model of cecal ligation and puncture (CLP), we analyzed the frequency and molecular signature of the T-cell immunoglobulin and ITIM domain (TIGIT)+ and TIGIT− Treg subsets, using flow cytometry and quantitative polymerase chain reaction. In addition, ST2−/− and signal transducer and activator of transcription 6 (STAT6)−/− mice were submitted to CLP or recombinant interleukin 33 (IL-33) treatment to investigate the mechanism whereby TIGIT+ Tregs differentiate during sepsis. Results Sepsis was marked by the sustained expansion of the highly suppressive TIGIT+ Treg subset, which expresses Helios, neuropilin 1, and high levels of Tnfrsf18 and Pdcd1 at 15 days after CLP. The increase in TIGIT+ Tregs was accompanied by higher susceptibility to nosocomial bacteria challenge, suggesting their association with post sepsis immunosuppression. Mechanistically, we found that the ST2 deletion abrogated the expansion of the TIGIT+ Treg subset during sepsis. Furthermore, treatment with recombinant IL-33 resulted in the expansion of TIGIT+ Tregs depending on the STAT6 and M2 macrophages. Conclusions These findings demonstrated that only the TIGIT+ Tregs remain stably expanded at the late phase of sepsis. Moreover, the expansion of TIGIT+ Tregs is dependent on the IL-33/ST2/STAT6/M2 macrophage axis.
- Published
- 2022
41. COVID‐19 bimodal clinical and pathological phenotypes
- Author
-
Sabrina S. Batah, Maíra N. Benatti, Li Siyuan, Wagner M. Telini, Jamile O. Barboza, Marcelo B. Menezes, Tales R. Nadai, Keyla S. G. Sá, Chirag M. Vaswani, Sahil Gupta, Dario S. Zamboni, Danilo T. Wada, Rodrigo T. Calado, Renê D. R. Oliveira, Paulo Louzada‐Junior, Maria Auxiliadora‐Martins, Flávio P. Veras, Larissa D. Cunha, Thiago M. Cunha, Rodrigo Luppino‐Assad, Marcelo L. Balancin, Sirlei S. Morais, Ronaldo B. Martins, Eurico Arruda, Fernando Chahud, Marcel Koenigkam Santos, Andrea A. Cetlin, Fernando Q. Cunha, Claudia dos Santos, Vera L. Capelozzi, Junya Fukuoka, Rosane Duarte Achcar, and Alexandre T. Fabro
- Subjects
Medicine (General) ,R5-920 ,Phenotype ,SARS-CoV-2 ,COVID-19 ,Humans ,Molecular Medicine ,Medicine (miscellaneous) ,Letter to Editor - Published
- 2022
42. Gasdermin D inhibition prevents multiple organ dysfunction during sepsis by blocking NET formation
- Author
-
Katiussia P Silva, David F. Colón, Valter Vinicius Silva Monteiro, Juliana E Toller-Kawahisa, Vanessa F. Borges, Fausto Almeida, Thiago M. Cunha, Helder I. Nakaya, Marcos C. Borges, José C. Alves-Filho, Verônica Soares Brauer, Dario S. Zamboni, Camila Meirelles de Souza Silva, Daniele C. Nascimento, Carlos W. S. Wanderley, Flávio P. Veras, Sabrina Setembre Batah, Luis Eduardo Alves Damasceno, Ana Letícia J Souza, Fabiane Sônego, Fernando Q. Cunha, Augusto V. Gonçalves, Paula B. Donate, Timna Varela Martins, Alexandre Todorovic Fabro, Antonio Edson Rocha Oliveira, Daniel Zoppi, Center for Research in Inflammatory Diseases, Universidade de São Paulo (USP), Universidade Estadual Paulista (UNESP), Pathology and Legal Medicine, and Sao Paulo
- Subjects
Male ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Multiple Organ Failure ,Immunology ,Acetaldehyde Dehydrogenase Inhibitors ,Disease ,Biochemistry ,Extracellular Traps ,Sepsis ,Phagocytes, Granulocytes, and Myelopoiesis ,Disulfiram ,medicine ,Animals ,Humans ,Cells, Cultured ,Aged ,business.industry ,Organ dysfunction ,Intracellular Signaling Peptides and Proteins ,Gasdermin D ,Cell Biology ,Hematology ,Neutrophil extracellular traps ,Middle Aged ,Phosphate-Binding Proteins ,medicine.disease ,Adoptive Transfer ,Mice, Inbred C57BL ,Pharmaceutical Preparations ,Female ,medicine.symptom ,business ,Gene Deletion ,medicine.drug - Abstract
Made available in DSpace on 2022-04-29T08:46:05Z (GMT). No. of bitstreams: 0 Previous issue date: 2021-12-23 Multiple organ dysfunction is the most severe outcome of sepsis progression and is highly correlated with a worse prognosis. Excessive neutrophil extracellular traps (NETs) are critical players in the development of organ failure during sepsis. Therefore, interventions targeting NET release would likely effectively prevent NET-based organ injury associated with this disease. Herein, we demonstrate that the pore-forming protein gasdermin D (GSDMD) is active in neutrophils from septic humans and mice and plays a crucial role in NET release. Inhibition of GSDMD with disulfiram or genic deletion abrogated NET formation, reducing multiple organ dysfunction and sepsis lethality. Mechanistically, we demonstrate that during sepsis, activation of the caspase-11/GSDMD pathway controls NET release by neutrophils during sepsis. In summary, our findings uncover a novel therapeutic use for disulfiram and suggest that GSDMD is a therapeutic target to improve sepsis treatment. Center for Research in Inflammatory Diseases Department of Biochemistry and Immunology Department of Pharmacology Department of Cellular and Molecular Biology and Pathogenic Bioagents Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto Institute of Biosciences Sao Paulo State University, Botucatu Pathology and Legal Medicine Department of Internal Medicine Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto Hospital Israelita Albert Einstein Sao Paulo Institute of Biosciences Sao Paulo State University, Botucatu
- Published
- 2021
43. Inflammasome activation and CCR2‐mediated monocyte‐derived dendritic cell recruitment restrict Legionella pneumophila infection
- Author
-
Marco A. Ataide, Graziele Z. Manin, Samuel S. Oliveira, Rhanoica O. Guerra, and Dario S. Zamboni
- Subjects
Immunology ,Immunology and Allergy - Abstract
Flagellin-induced NAIP/NLRC4 inflammasome activation and pyroptosis are critical events restricting Legionella pneumophila infection. However, the cellular and molecular dynamics of the in vivo responses against this bacterium are still unclear. We have found temporal coordination of two independent innate immunity pathways in controlling Legionella infection, the inflammasome activation and the CCR2-mediated Mo-DC recruitment. Inflammasome activation was an important player at the early stage of infection by lowering the numbers of bacteria for an efficient bacterial clearance conferred by the Mo-DC at the late stage of the infection. Mo-DC emergence highly depended on CCR2-signaling and dispensed inflammasome activation and pyroptosis. Also, Mo-DC compartment did not rely on the inflammasome machinery to deliver proper immune responses and was the most abundant cytokine-producing among the monocyte-derived cells in the infected lung. Importantly, when the CCR2- and NLRC4-dependent axes of response were simultaneously ablated, we observed an aggravated bacterial burden in the lung of infected mice. Taken together, we showed that inflammasome activation and CCR2-mediated immune response interplay in distinct pathways to restrict pulmonary bacterial infection. These findings extend our understanding of the in vivo integration and cooperation of different innate immunity arms in controlling infectious agents.
- Published
- 2022
44. Lipid droplet accumulation occurs early following Salmonella infection and contributes to intracellular bacterial survival and replication
- Author
-
Filipe S. Pereira-Dutra, Patrícia T. Bozza, Warrison A. Andrade, Felipe Ferraro-Moreira, Tamires Cunha-Fernandes, Marcelo T. Bozza, Julia da Cunha Santos, Taynná C Goltara-Gomes, Matheus Andrade Rajão, Dario S. Zamboni, Ellen Kiarely Souza, and Elisa Beatriz Prestes
- Subjects
Salmonella ,Intracellular parasite ,media_common.quotation_subject ,Macrophages ,Lipid metabolism ,Lipid Droplets ,Biology ,medicine.disease_cause ,Lipid Metabolism ,Microbiology ,Cell biology ,Type three secretion system ,Lipid droplet ,Salmonella Infections ,medicine ,Type III Secretion Systems ,Humans ,Internalization ,Molecular Biology ,Intracellular ,Diacylglycerol kinase ,media_common - Abstract
Salmonellosis is a public health problem caused by Salmonella sp., a highly adapted facultative intracellular pathogen. After internalization, Salmonella sp. manipulates several host processes, mainly through the activation of the type III secretion system (T3SS), including modification of host lipid metabolism and lipid droplet (LD) accumulation. LDs are dynamic and complex lipid-rich organelles involved in several cellular processes. The present study investigated the mechanism involved in LD biogenesis in Salmonella-infected macrophages and its role in bacterial pathogenicity. Here, we reported that S. Typhimurium induced a rapid time-dependent increase of LD formation in macrophages. The LD biogenesis was demonstrated to depend on Salmonella's viability and SPI1-related T3SS activity, with the participation of Toll-Like Receptor (TLR) signaling. We also observed that LD accumulation occurs through TLR2 dependent signaling and is counter-regulated by TLR4. Lastly, the pharmacological modulation of LD formation by inhibiting diacylglycerol O-acyltransferase 1 (DGAT1) and cytosolic phospholipase A2 (cPLA2) significantly reduced the intracellular bacterial proliferation and impaired the prostaglandin E2 (PGE2 ) synthesis. Collectively, our data suggest the role of LDs on S. Typhimurium intracellular survival and replication in macrophages. This data set provides new perspectives for future investigations about LDs in host-pathogen interaction.
- Published
- 2021
45. Chikungunya Virus Exposure Partially Cross-Protects against Mayaro Virus Infection in Mice
- Author
-
Dario S. Zamboni, Luiz Tadeu Moraes Figueiredo, Luiz Gustavo de Almeida, Sílvio Roberto Consonni, Luiza Antunes de Castro-Jorge, William Marciel de Souza, Italo A. Castro, Marcílio Jorge Fumagalli, and Renan V. H. de Carvalho
- Subjects
Secondary infection ,viruses ,Cross Protection ,Immunology ,Alphavirus ,Semliki Forest virus ,medicine.disease_cause ,Antibodies, Viral ,Microbiology ,Virus ,Mice ,Immune system ,Immunity ,Virology ,medicine ,Animals ,Chikungunya ,Epidemics ,Inflammation ,biology ,Alphavirus Infections ,virus diseases ,Viral Load ,biology.organism_classification ,Mice, Inbred C57BL ,Disease Models, Animal ,Insect Science ,SURTOS DE DOENÇAS ,Chikungunya Fever ,Pathogenesis and Immunity ,Female ,Viral load ,Chikungunya virus - Abstract
Chikungunya virus (CHIKV) and Mayaro virus (MAYV) are closely related members of the Semliki Forest virus antigenic complex classified as belonging to the genus Alphavirus of the family Togaviridae. These viruses cause human disease, with sudden fever and joint inflammation that can persist for long periods. CHIKV is the causative agent of large outbreaks worldwide, and MAYV infection represents a growing public health concern in Latin America, causing sporadic cases and geographically limited outbreaks. Considering the relationship between CHIKV and MAYV, the present study aimed to evaluate if preexisting CHIKV immunity protects against MAYV infection. Immunocompetent C57BL/6 mice were intraperitoneally infected with CHIKV and, 4 weeks later, they were infected with MAYV in their hind paw. We observed that the preexistence of CHIKV immunity conferred partial cross-protection against secondary MAYV infection, reducing disease severity, tissue viral load, and histopathological scores. Interestingly, CHIKV antibodies from humans and mice showed low cross-neutralization to MAYV, but neutralizing activity significantly increased after secondary infection. Furthermore, depletion of adaptive immune cells (CD4(+) T, CD8(+) T, and CD19(+) B cells) did not alter the cross-protection phenotype, suggesting that distinct cell subsets or a combination of adaptive immune cells stimulated by CHIKV are responsible for the partial cross-protection against MAYV. The reduction of proinflammatory cytokines, such as interferon gamma (IFN-γ), in animals secondarily infected by MAYV, suggests a role for innate immunity in cross-protection. Our findings shed light on how preexisting immunity to arthritogenic alphaviruses may affect secondary infection, which may further develop relevant influence in disease outcome and viral transmission. IMPORTANCE Mosquito-borne viruses have a worldwide impact, especially in tropical climates. Chikungunya virus has been present mostly in developing countries, causing millions of infections, while Mayaro virus, a close relative, has been limited to the Caribbean and tropical regions of Latin America. The potential emergence and spread of Mayaro virus to other high-risk areas have increased the scientific community’s attention to an imminent worldwide epidemic. Here, we designed an experimental protocol of chikungunya and Mayaro virus mouse infection, which develops a measurable and quantifiable disease that allows us to make inferences about potential immunological effects during secondary virus infection. Our results demonstrate that previous chikungunya virus infection is able to reduce the severity of clinical outcomes during secondary Mayaro infection. We provide scientific understanding of immunological features during secondary infection with the closely related virus, thus assisting in better comprehending viral transmission and the pathological outcome of these diseases.
- Published
- 2021
46. Keeping the host alive - lessons from obligate intracellular bacterial pathogens
- Author
-
Hayley J. Newton, Robson Kriiger Loterio, and Dario S. Zamboni
- Subjects
Microbiology (medical) ,Virulence Factors ,Cell ,Intracellular Space ,Virulence ,Bacterial Physiological Phenomena ,Phagocytosis ,Species Specificity ,medicine ,Immunology and Allergy ,Animals ,Humans ,Microbial Viability ,General Immunology and Microbiology ,biology ,Obligate ,Cell Death ,Host (biology) ,General Medicine ,Coxiella burnetii ,biology.organism_classification ,Cell biology ,Oxidative Stress ,Infectious Diseases ,medicine.anatomical_structure ,Rickettsia ,Host-Pathogen Interactions ,Disease Susceptibility ,Signal transduction ,Lysosomes ,Intracellular ,Biomarkers - Abstract
Mammals have evolved sophisticated host cell death signaling pathways as an important immune mechanism to recognize and eliminate cell intruders before they establish their replicative niche. However, intracellular bacterial pathogens that have co-evolved with their host have developed a multitude of tactics to counteract this defense strategy to facilitate their survival and replication. This requires manipulation of pro-death and pro-survival host signaling pathways during infection. Obligate intracellular bacterial pathogens are organisms that absolutely require an eukaryotic host to survive and replicate, and therefore they have developed virulence factors to prevent diverse forms of host cell death and conserve their replicative niche. This review encapsulates our current understanding of these host–pathogen interactions by exploring the most relevant findings of Anaplasma spp., Chlamydia spp., Rickettsia spp. and Coxiella burnetii modulating host cell death pathways. A detailed comprehension of the molecular mechanisms through which these obligate intracellular pathogens manipulate regulated host cell death will not only increase the current understanding of these difficult-to-study pathogens but also provide insights into new tools to study regulated cell death and the development of new therapeutic approaches to control infection.
- Published
- 2021
47. COVID-19 Bimodal Clinical and Pathological Phenotypes
- Author
-
Marcelo Bezerra de Menezes, Chirag M. Vaswani, Junya Fukuoka, Rodrigo Luppino-Assad, Marcelo Luiz Balancin, Sirlei Antunes de Morais, Jamile Barbosa, Sahil Gupta, Thiago M. Cunha, Dario S. Zamboni, Vera Luiza Capelozzi, Sabrina Setembre Batah, Fernando Chahud, Tales Rubens de Nadai, Danilo Wada, Maira N. Benatti, Paulo Louzada-Junior, Claudia C. dos Santos, Rodrigo T. Calado, Marcel Koenigkam-Santos, Wagner Telini, Maria Auxiliadora-Martins, Alexandre Todorovic Fabro, Fernando Q. Cunha, Keyla S, Larissa D. Cunha, Renê Donizeti Ribeiro de Oliveira, Eurico Arruda, Ronaldo Teixeira Martins, Andrea Cetlin, Rosane Duarte-Achcar, Flávio P. Veras, and Li Siyuan
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Autopsy ,Pulmonary compliance ,medicine.disease ,Immunofluorescence ,Stain ,Medicine ,Immunohistochemistry ,Thrombus ,business ,Pathological ,Viral load - Abstract
BackgroundPatients with coronavirus disease-2019 (COVID-19) present varying clinical complications. Different viral load and host response related to genetic and immune background are probably the reasons for these differences. We aimed to sought clinical and pathological correlation that justifies the different clinical outcomes among COVID-19 autopsies cases.MethodsMinimally invasive autopsy was performed on forty-seven confirmed COVID-19 patients from May-July, 2020. Electronic medical record of all patients was collected and a comprehensive histopathological evaluation was performed. Immunohistochemistry, immunofluorescence, special stain, western blotting and post-mortem real-time reverse transcriptase polymerase chain reaction on fresh lung tissue were performed.ResultsWe show that 5/47 (10,6%) patients present a progressive decline in oxygenation index for acute respiratory distress syndrome (PaO2/FiO2ratio), low compliance levels, interstitial fibrosis, high α-SMA+ cells/protein expression, high collagens I/III deposition and NETs(P2/FiO2ratio, high pulmonary compliance levels, preserved elastic framework, increase thrombus formation and high platelets and D-dimer levels at admission (PConclusionsWe believe that categorization of patients based on these two phenotypes can be used to develop prognostic tools and potential therapies since the PaO2/FiO2ratio variation and D-dimer levels correlate with the underlying fibrotic or thrombotic pathologic process, respectively; which may indicate possible clinical outcome of the patient.
- Published
- 2021
48. Disease severity and mortality can be independently regulated in a mouse model of experimental graft versus host disease.
- Author
-
Rômulo G Galvani, Ramon Lemos, Rômulo B Areal, Pollyanna A Salvador, Dario S Zamboni, João Luiz M Wanderley, and Adriana Bonomo
- Subjects
Medicine ,Science - Abstract
Graft versus host disease (GVHD) is the major limitation of allogeneic hematopoietic stem cell transplantation (HSCT) presenting high mortality and morbidity rates. However, the exact cause of death is not completely understood and does not correlate with specific clinical and histological parameters of disease. Here we show, by using a semi-allogeneic mouse model of GVHD, that mortality and morbidity can be experimentally separated. We injected bone marrow-derived dendritic cells (BMDC) from NOD2/CARD15-deficient donors into semi-allogeneic irradiated chimaeras and observed that recipients were protected from death. However, no protection was observed regarding clinical or pathological scores up to 20 days after transplantation. Protection from death was associated with decreased bacterial translocation, faster hematologic recovery and epithelial integrity maintenance despite mononuclear infiltration at day 20 post-GVHD induction with no skew towards different T helper phenotypes. The protected mice recovered from aGVHD and progressively reached scores compatible with healthy animals. Altogether, our data indicate that severity and mortality can be separate events providing a model to study transplant-related mortality.
- Published
- 2015
- Full Text
- View/download PDF
49. Perioperative complications after radical cystectomy for bladder cancer: an international multicenter collaboration
- Author
-
C. Lonati, L. Afferi, E. Di Trapani, O. De Cobelli, F. Mondini, S. Zamboni, N. Suardi, C. Simeone, G. Basile, M. Bandini, A. Briganti, F. Montorsi, S. Albisinni, G. Simone, R. Mastroianni, F. Soria, G. Marra, P. Gontero, C. Mir, R. Sanchez-Salas, M. Roumiguié, P.I. Ornaghi, A. Panunzio, A. Gozzo, A. Tafuri, M.A. Cerruto, A. Antonelli, K. Hendricksen, A. Mattei, J. Teoh, R. Contieri, R. Hurle, P. Umari, J. Kelly, A. Olivero, P. Dell’Oglio, S. Secco, A. Galfano, S. Francavilla, A. Peroni, E. Xylinas, W. Krajewski, M. Borghesi, G. Mantica, C. Terrone, M. Rouprêt, E. Laukhtina, G. Ploussard, S. Shariat, and M. Moschini
- Subjects
Urology - Published
- 2022
50. Independent predictors and patterns of early and late recurrence after radical cystectomy for bladder cancer: a multicenter collaboration
- Author
-
C. Lonati, R. Carando, A. Martini, C. Simeone, N. Suardi, S. Zamboni, G. Simone, L. Afferi, C.D. Fankhauser, A. Mattei, M. Álvarez-Maestro, A. Briganti, F. Montorsi, and M. Moschini
- Subjects
Urology - Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.