1
|
Novaes RD, Souza-E-Leite EG, Silva TD, Caetano-da-Silva JE, Caldas IS, Souza RLM, Marques MJ, Gonçalves RV. Pharmacological blockade of infection chronification modulates oxy-inflammation and prevents the activation of stress-induced premature senescence markers in schistosomiasis. Microb Pathog 2025; 199:107264. [PMID: 39732412 DOI: 10.1016/j.micpath.2024.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Chronic inflammation, oxidative stress, and DNA damage are observed in schistosomiasis and premature aging. However, the potential of these events to trigger stress-induced premature senescence (SIPS) throughout schistosomiasis progression remains overlooked, especially in response to the first-line pharmacological treatment. Thus, we investigated the relationship between oxidative stress and SIPS sentinel markers in untreated Schistosoma mansoni-infected mice and those receiving praziquantel (Pz)-based reference treatment. Swiss mice were randomized into 5 groups: uninfected (followed by 60- and 180-days post-infection), acutely (60 days) and chronically (180 days) infected untreated, and infected treated with Pz followed until 180 days. Our results indicated that infection chronification was accompanied by the worsening of hepatic granulomatous inflammation, increased number of granulomas, IL-4, TGF-β, reactive oxygen species (ROS) levels, fibrosis, hepatocytes DNA damage, upregulation in SA-β-gal activity, p16 and p21 gene expression, and hepatocytes proliferation down-regulation in the absence of telomeric shortening. These abnormalities were blocked by Pz treatment, which prevented infection chronification and the decline in hepatocytes proliferative potential, stimulating granulomatous inflammation resolution. Taken together, our findings provide the evidence that progressive fibrosis, sustained production of high ROS levels, marked DNA damage and decline in p16 and p21 expression are associated with hepatocytes replication attenuation in the chronic phase of S. mansoni infection. Thus, pharmacological blockade of infection and granulomatous inflammation is essential to prevent these premature senescence markers associated with hepatocytes replicative disorders, stimulating liver regeneration in schistosomiasis mansoni.
Collapse
Affiliation(s)
- Rômulo D Novaes
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Departamento de Biologia Animal (DBA), Programa de Pós-Graduação em Biologia Animal (PPGBA), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil.
| | - Elda G Souza-E-Leite
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Thiago D Silva
- Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - José Edson Caetano-da-Silva
- Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Ivo S Caldas
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Raquel L M Souza
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Marcos J Marques
- Instituto de Ciências Biomédicas (ICB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas (PPGCB), Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, 37130-001, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Departamento de Biologia Animal (DBA), Programa de Pós-Graduação em Biologia Animal (PPGBA), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil
| |
Collapse
|
2
|
Caetano-da-Silva JE, Gonçalves-Santos E, Domingues ELBC, Caldas IS, Lima GDA, Diniz LF, Gonçalves RV, Novaes RD. The mitochondrial uncoupler 2,4-dinitrophenol modulates inflammatory and oxidative responses in Trypanosoma cruzi-induced acute myocarditis in mice. Cardiovasc Pathol 2024; 72:107653. [PMID: 38740356 DOI: 10.1016/j.carpath.2024.107653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
By uncoupling oxidative phosphorylation, 2,4-dinitrophenol (DNP) attenuates reactive oxygen species (ROS) biosynthesis, which are known to aggravate infectious myocarditis in Chagas disease. Thus, the impact of DNP-based chemotherapy on Trypanosoma cruzi-induced acute myocarditis was investigated. C56BL/6 mice uninfected and infected untreated and treated daily with 100 mg/kg benznidazole (Bz, reference drug), 5 and 10 mg/kg DNP by gavage for 11 days after confirmation of T. cruzi infection were investigated. Twenty-four hours after the last treatment, the animals were euthanized and the heart was collected for microstructural, immunological and biochemical analyses. T. cruzi inoculation induced systemic inflammation (e.g., cytokines and anti-T. cruzi IgG upregulation), cardiac infection (T. cruzi DNA), oxidative stress, inflammatory infiltrate and microstructural myocardial damage in untreated mice. DNP treatment aggravated heart infection and microstructural damage, which were markedly attenuated by Bz. DNP (10 mg/kg) was also effective in attenuating ROS (total ROS, H2O2, and O2-), nitric oxide (NO), lipid (malondialdehyde - MDA) and protein (protein carbonyl - PCn) oxidation, TNF, IFN-γ, IL-10, and MCP-1/CCL2, anti-T. cruzi IgG, cardiac troponin I levels, as well as inflammatory infiltrate and cardiac damage in T. cruzi-infected mice. Our findings indicate that DNP aggravated heart infection and microstructural cardiomyocytes damage in infected mice. These responses were related to the antioxidant and anti-inflammatory properties of DNP, which favors infection by weakening the pro-oxidant and pro-inflammatory protective mechanisms of the infected host. Conversely, Bz-induced cardioprotective effects combined effective anti-inflammatory and antiparasitic responses, which protect against heart infection, oxidative stress, and microstructural damage in Chagas disease.
Collapse
Affiliation(s)
- José Edson Caetano-da-Silva
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Elisa L B C Domingues
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Ivo S Caldas
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Graziela D A Lima
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Lívia F Diniz
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil; Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Liz Belli Cassa Domingues E, Gonçalves-Santos E, Santana Caldas I, Vilela Gonçalves R, Caetano-da-Silva JE, Cardoso Santos E, Mól Pelinsari S, Figueiredo Diniz L, Dias Novaes R. Identification of host antioxidant effectors as thioridazine targets: Impact on cardiomyocytes infection and Trypanosoma cruzi-induced acute myocarditis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167264. [PMID: 38806073 DOI: 10.1016/j.bbadis.2024.167264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Phenothiazines inhibit antioxidant enzymes in trypanosomatids. However, potential interferences with host cell antioxidant defenses are central concerns in using these drugs to treat Trypanosoma cruzi-induced infectious myocarditis. Thus, the interaction of thioridazine (TDZ) with T. cruzi and cardiomyocytes antioxidant enzymes, and its impact on cardiomyocytes and cardiac infection was investigated in vitro and in vivo. Cardiomyocytes and trypomastigotes in culture, and mice treated with TDZ and benznidazole (Bz, reference antiparasitic drug) were submitted to microstructural, biochemical and molecular analyses. TDZ was more cytotoxic and less selective against T. cruzi than Bz in vitro. TDZ-pretreated cardiomyocytes developed increased infection rate, reactive oxygen species (ROS) production, lipid and protein oxidation; similar catalase (CAT) and superoxide dismutase (SOD) activity, and reduced glutathione's (peroxidase - GPx, S-transferase - GST, and reductase - GR) activity than infected untreated cells. TDZ attenuated trypanothione reductase activity in T. cruzi, and protein antioxidant capacity in cardiomyocytes, making these cells more susceptible to H2O2-based oxidative challenge. In vivo, TDZ potentiated heart parasitism, total ROS production, myocarditis, lipid and protein oxidation; as well as reduced GPx, GR, and GST activities compared to untreated mice. Benznidazole decreased heart parasitism, total ROS production, heart inflammation, lipid and protein oxidation in T. cruzi-infected mice. Our findings indicate that TDZ simultaneously interact with enzymatic antioxidant targets in cardiomyocytes and T. cruzi, potentiating the infection by inducing antioxidant fragility and increasing cardiomyocytes and heart susceptibility to parasitism, inflammation and oxidative damage.
Collapse
Affiliation(s)
- Elisa Liz Belli Cassa Domingues
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Ivo Santana Caldas
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Reggiani Vilela Gonçalves
- Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil; Programa de Pós-Graduação em Biologia Celular e Estrutural, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - José Edson Caetano-da-Silva
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Eliziária Cardoso Santos
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina 39100-000, Minas Gerais, Brazil
| | - Silvania Mól Pelinsari
- Programa de Pós-Graduação em Biologia Celular e Estrutural, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Lívia Figueiredo Diniz
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Rômulo Dias Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Perera DJ, Koger-Pease C, Paulini K, Daoudi M, Ndao M. Beyond schistosomiasis: unraveling co-infections and altered immunity. Clin Microbiol Rev 2024; 37:e0009823. [PMID: 38319102 PMCID: PMC10938899 DOI: 10.1128/cmr.00098-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kayla Paulini
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Mohamed Daoudi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
5
|
Lozano KJG, Gonçalves Santos E, Vilas Boas DF, Oliveira RRG, Diniz LF, Benedetti MD, Carneiro CM, C Bandeira L, Faria G, Gonçalves RV, Novaes RD, Caldas S, Caldas IS. Schistosoma mansoni co-infection modulates Chagas disease development but does not impair the effect of benznidazole-based chemotherapy. Int Immunopharmacol 2024; 128:111467. [PMID: 38211479 DOI: 10.1016/j.intimp.2023.111467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024]
Abstract
The adequate management of parasite co-infections represents a challenge that has not yet been overcome, especially considering that the pathological outcomes and responses to treatment are poorly understood. Thus, this study aimed to evaluate the impact of Schistosoma mansoni infection on the efficacy of benznidazole (BZN)-based chemotherapy in Trypanosoma cruzi co-infected mice. BALB/c mice were maintained uninfected or co-infected with S. mansoni and T. cruzi, and were untreated or treated with BZN. Body weight, mortality, parasitemia, cardiac parasitism, circulating cytokines (Th1/Th2/Th17); as well as heart, liver and intestine microstructure were analyzed. The parasitemia peak was five times higher and myocarditis was more severe in co-infected than T. cruzi-infected mice. After reaching peak, parasitemia was effectively controlled in co-infected animals. BZN successfully controlled parasitemia in both co-infected and T. cruzi-infected mice and improved body mass, cardiac parasitism, myocarditis and survival in co-infected mice. Co-infection dampened the typical cytokine response to either parasite, and BZN reduced anti-inflammatory cytokines in co-infected mice. Despite BZN normalizing splenomegaly and liver cellular infiltration, it exacerbated hepatomegaly in co-infected mice. Co-infection or BZN exerted no effect on hepatic granulomas, but increased pulmonary and intestinal granulomas. Marked granulomatous inflammation was identified in the small intestine of all schistosomiasis groups. Taken together, our findings indicate that BZN retains its therapeutic efficacy against T. cruzi infection even in the presence of S. mansoni co-infection, but with organ-specific repercussions, especially in the liver.
Collapse
Affiliation(s)
- Kelly J G Lozano
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Elda Gonçalves Santos
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Diego F Vilas Boas
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Raphaela R G Oliveira
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Lívia F Diniz
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Monique D Benedetti
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Cláudia M Carneiro
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University, Ouro Preto 35400-000, MG, Brazil
| | - Lorena C Bandeira
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University, Ouro Preto 35400-000, MG, Brazil
| | - Gilson Faria
- Department of Research and Development., Ezequiel Dias Foundation, 30510-010, Belo Horizonte, MG, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | - Rômulo D Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-000, MG, Brazil
| | - Sérgio Caldas
- Department of Research and Development., Ezequiel Dias Foundation, 30510-010, Belo Horizonte, MG, Brazil
| | - Ivo S Caldas
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil.
| |
Collapse
|
6
|
de Souza G, Teixeira SC, Fajardo Martínez AF, Silva RJ, Luz LC, de Lima Júnior JP, Rosini AM, dos Santos NCL, de Oliveira RM, Paschoalino M, Barbosa MC, Alves RN, Gomes AO, da Silva CV, Ferro EAV, Barbosa BF. Trypanosoma cruzi P21 recombinant protein modulates Toxoplasma gondii infection in different experimental models of the human maternal-fetal interface. Front Immunol 2023; 14:1243480. [PMID: 37915581 PMCID: PMC10617204 DOI: 10.3389/fimmu.2023.1243480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction Toxoplasma gondii is the etiologic agent of toxoplasmosis, a disease that affects about one-third of the human population. Most infected individuals are asymptomatic, but severe cases can occur such as in congenital transmission, which can be aggravated in individuals infected with other pathogens, such as HIV-positive pregnant women. However, it is unknown whether infection by other pathogens, such as Trypanosoma cruzi, the etiologic agent of Chagas disease, as well as one of its proteins, P21, could aggravate T. gondii infection. Methods In this sense, we aimed to investigate the impact of T. cruzi and recombinant P21 (rP21) on T. gondii infection in BeWo cells and human placental explants. Results Our results showed that T. cruzi infection, as well as rP21, increases invasion and decreases intracellular proliferation of T. gondii in BeWo cells. The increase in invasion promoted by rP21 is dependent on its binding to CXCR4 and the actin cytoskeleton polymerization, while the decrease in proliferation is due to an arrest in the S/M phase in the parasite cell cycle, as well as interleukin (IL)-6 upregulation and IL-8 downmodulation. On the other hand, in human placental villi, rP21 can either increase or decrease T. gondii proliferation, whereas T. cruzi infection increases T. gondii proliferation. This increase can be explained by the induction of an anti-inflammatory environment through an increase in IL-4 and a decrease in IL-6, IL-8, macrophage migration inhibitory factor (MIF), and tumor necrosis factor (TNF)-α production. Discussion In conclusion, in situations of coinfection, the presence of T. cruzi may favor the congenital transmission of T. gondii, highlighting the importance of neonatal screening for both diseases, as well as the importance of studies with P21 as a future therapeutic target for the treatment of Chagas disease, since it can also favor T. gondii infection.
Collapse
Affiliation(s)
- Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Luana Carvalho Luz
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Joed Pires de Lima Júnior
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Natália Carine Lima dos Santos
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rafael Martins de Oliveira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marina Paschoalino
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Matheus Carvalho Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rosiane Nascimento Alves
- Department of Agricultural and Natural Science, Universidade do Estado de Minas Gerais, Ituiutaba, MG, Brazil
| | - Angelica Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Claudio Vieira da Silva
- Laboratory of Trypanosomatids, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
7
|
Jones KM, Zhan B, Ernste KJ, Villar MJ, Bisht N, Nguyen D, Chang LY, Poveda C, Robinson GJ, Trivedi AJ, Hofferek CJ, Decker WK, Konduri V. Immunomodulatory proteins from hookworms reduce cardiac inflammation and modulate regulatory responses in a mouse model of chronic Trypanosoma cruzi infection. FRONTIERS IN PARASITOLOGY 2023; 2:1244604. [PMID: 38239430 PMCID: PMC10795693 DOI: 10.3389/fpara.2023.1244604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/19/2023] [Indexed: 01/22/2024]
Abstract
Introduction Hookworms are parasitic helminths that secrete a variety of proteins that induce anti-inflammatory immune responses, stimulating increased CD4 + Foxp3+ regulatory T cells and IL-10 production. Hookworm-derived recombinant proteins AIP-1 and AIP-2 have been shown to reduce inflammation in mouse models of inflammatory bowel disease and inflammatory airway disease by inducing CD4+Foxp3+ cells and IL-10 production. In contrast, chronic infection with the protozoal parasite Trypanosoma cruzi, the causative agent of Chagas disease, leads to chronic inflammation in tissues. Persistence of the parasites in tissues drives chronic low-grade inflammation, with increased infiltration of inflammatory cells into the heart, accompanied by increased production of inflammatory cytokines. There are no current antiparasitic drugs that effectively reduce or prevent chronic myocarditis caused by the onset of Chagas disease, thus new therapies are urgently needed. Therefore, the impact of AIP-1 and AIP-2 on myocarditis was investigated in a mouse model of chronic T. cruzi infection. Methods Female BALB/c mice infected with bioluminescent T. cruzi H1 strain trypomastigotes for 70 days were treated once daily for 7 days with 1mg/kg AIP-1 or AIP-2 protein by intraperitoneal injection. Control mice were left untreated or treated once daily for 14 days with 25mg/kg aspirin in drinking water. At 84 days of infection, splenocytes, cardiac tissue and serum were collected for evaluation. Results Treatment with both AIP-1 and AIP-2 proteins significantly reduced cardiac cellular infiltration, and reduced cardiac levels of IFNγ, IL-6 and IL-2. AIP-2 treatment reduced cardiac expression of COX-2. Further, while incubation with AIP-1 and AIP-2 proteins did not induce a significant upregulation of an immunoregulatory phenotype in dendritic cells (DC), there was a modest upregulation of CD11c +CD11b+MHCII+SIRPα+ expression, suggesting a regulatory phenotype. Ex-vivo stimulation of splenocytes from the treatment groups with AIP-1 loaded DC induced reduced levels of cytotoxic and pro-inflammatory T cells, stimulation with AIP-2 loaded DC specifically induced enhanced levels of CD4+CD25+Foxp3+ regulatory T cells among treatment groups. Discussion All in vivo and in vitro results demonstrate that hookworm-derived AIP-1 and AIP-2 proteins reduce T. cruzi induced cardiac inflammation, possibly through multiple anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Kathryn M. Jones
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Bin Zhan
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Keenan J. Ernste
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Maria Jose Villar
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Nalini Bisht
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Duc Nguyen
- Center for Comparative Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Li-Yen Chang
- Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Cristina Poveda
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Gonteria J. Robinson
- Molecular & Human Genetics Department, Baylor College of Medicine, Houston, TX, United States
| | - Akshar J. Trivedi
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Colby J. Hofferek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - William K. Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer, Baylor College of Medicine, Houston, TX, United States
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
| | - Vanaja Konduri
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
8
|
Gonçalves-Santos E, Caldas IS, Fernandes VÂ, Franco LL, Pelozo MF, Feltrim F, Maciel JS, Machado JVC, Gonçalves RV, Novaes RD. Pharmacological potential of new metronidazole/eugenol/dihydroeugenol hybrids against Trypanosoma cruzi in vitro and in vivo. Int Immunopharmacol 2023; 121:110416. [PMID: 37295025 DOI: 10.1016/j.intimp.2023.110416] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/11/2023]
Abstract
AIMS From well-delimited immunomodulatory, redox and antimicrobial properties; metronidazole and eugenol were used as structural platforms to assembly two new molecular hybrids (AD06 and AD07), whose therapeutic relevance was analyzed on T. cruzi infection in vitro and in vivo. METHODS Non-infected, T. cruzi-infected H9c2 cardiomyocytes, and mice non-treated and treated with vehicle, benznidazole (Bz - reference drug), AD06 and AD07 were investigated. Parasitological, prooxidant, antioxidant, microstructural, immunological, and hepatic function markers were analyzed. RESULTS Our findings indicated that in addition to having a direct antiparasitic effect on T. cruzi, metronidazole/eugenol hybrids (especially AD07) attenuated cellular parasitism, reactive species biosynthesis and oxidative stress in infected cardiomyocytes in vitro. Although AD06 and AD07 exerted no relevant impact on antioxidant enzymes activity (CAT, SOD, GR and GPx) in host cells, these drugs (especially AD07) attenuated trypanothione reductase activity in T. cruzi, which increased parasite's susceptibility to in vitro pro-oxidant challenge. AD06 and AD07 were well tolerated and do not determine humoral response suppression, mortality (100 % survival) or hepatotoxicity in mice, as indicated by transaminases plasma levels. AD07 also induced relevant in vivo antiparasitic and cardioprotective effects, attenuating parasitemia, cardiac parasite load and myocarditis in T. cruzi-infected mice. Although this cardioprotective response is potentially related to AD07 antiparasitic effect, a direct anti-inflammatory potential of this molecular hybrid cannot be ruled out. CONCLUSION Taken together, our findings indicated that the new molecular hybrid AD07 stood out as a potentially relevant candidate for the development of new, safe and more effective drug regimens for T. cruzi infection treatment.
Collapse
Affiliation(s)
- Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Ivo S Caldas
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Valquiria  Fernandes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Lucas L Franco
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Mônica F Pelozo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Fernando Feltrim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Juliana S Maciel
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Medicamentos, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Jose Vaz C Machado
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Departamento de Biologia Animal, Programa de Pós-Graduação em Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil.
| |
Collapse
|
9
|
Nogueira SS, Souza MA, Santos EC, Caldas IS, Gonçalves RV, Novaes RD. Oxidative stress, cardiomyocytes senescence and contractile dysfunction in in vitro and in vivo experimental models of Chagas disease. Acta Trop 2023:106950. [PMID: 37211152 DOI: 10.1016/j.actatropica.2023.106950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/13/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
AIMS The relationship between redox imbalance and cardiovascular senescence in infectious myocarditis is unknown. Thus, the aim of this study was to investigate whether cardiomyocytes parasitism, oxidative stress and contractile dysfunction can be correlated to senescence-associated β-galactosidase (SA-β-Gal) activity in Trypanosoma cruzi-infection in vitro and in vivo. METHODS Uninfected, T. cruzi-infected untreated and benznidazole (BZN)-treated H9c2 cardiomyocytes and rats were investigated. Parasitological, prooxidant, antioxidant, microstructural, and senescence-associated markers were quantified in vitro and in vivo. RESULTS T. cruzi infection triggered intense cardiomyocytes parasitism in vitro and in vivo, which was accompanied by reactive oxygen species (ROS) upregulation, lipids, proteins and DNA oxidation in cardiomyocytes and cardiac tissue. Oxidative stress was parallel to microstructural cell damage (e.g., increased cardiac toponin I levels) and contractile dysfunction in cardiomyocytes in vitro and in vivo, whose severity accompanied a premature cellular senescence-like phenotype revealed by increased senescence-associated β-galactosidase (SA-β-Gal) activity and DNA oxidation (8-OHdG). Cellular parasitism (e.g., infection rate and parasite load), myocarditis and T. cruzi-induced prooxidant responses were attenuated by early BZN administration to interrupt the progression of T. cruzi infection, protecting against SA-β-gal-based premature cellular senescence, microstructural damage and contractile deterioration in cardiomyocytes from T. cruzi-infected animals. CONCLUSION Our findings indicated that cell parasitism, redox imbalance and contractile dysfunction were correlated to SA-β-Gal-based cardiomyocytes premature senescence in acute T. cruzi infection. Therefore, in addition to controlling parasitism, inflammation and oxidative stress; inhibiting cardiomyocytes premature senescence should be further investigated as an additional target of specific Chagas disease therapeutics.
Collapse
Affiliation(s)
- Silas Santana Nogueira
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil; Instituto Federal do Sul de Minas Gerais, Pouso Alegre, 37560-250, Minas Gerais, Brazil
| | - Matheus Augusto Souza
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil
| | - Eliziária Cardoso Santos
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brazil, 39100-000, Minas Gerais, Brazil
| | - Ivo Santana Caldas
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil
| | - Reggiani Vilela Gonçalves
- Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa, 36570-900, Minas Gerais, Brazil
| | - Rômulo Dias Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, 37130-000, Minas Gerais, Brazil.
| |
Collapse
|
10
|
Souza MA, Gonçalves-Santos E, Gonçalves RV, Santos EC, Campos CC, Marques MJ, Souza RL, Novaes RD. Doxycycline hyclate stimulates inducible nitric oxide synthase and arginase imbalance, potentiating inflammatory and oxidative lung damage in schistosomiasis. Biomed J 2022; 45:857-869. [PMID: 34971826 PMCID: PMC9795368 DOI: 10.1016/j.bj.2021.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 11/19/2021] [Accepted: 12/21/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We investigated the relationship between inducible nitric oxide synthase (iNOS) and arginase pathways, cytokines, macrophages, oxidative damage and lung granulomatous inflammation in S. mansoni-infected and doxycycline-treated mice. METHODS Swiss mice were randomized in four groups: (i) uninfected, (ii) infected with S. mansoni, (iii) infected + 200 mg/kg praziquantel (Pzt), (iv) and (v) infected + 5 and 50 mg/kg doxycycline. Pzt (reference drug) was administered in a single dose and doxycycline for 60 days. RESULTS S. mansoni-infection determined extensive lung inflammation, marked recruitment of M2 macrophages, cytokines (IL-4, IL-5, IFN-γ, TNF-α) upregulation, intense eosinophil peroxidase (EPO) levels, arginase expression and activity, reduced iNOS expression and nitric oxide (NO) production. The higher dose of doxycycline aggravated lung granulomatous inflammation, downregulating IL-4 levels and M2 macrophages recruitment, and upregulating iNOS expression, EPO, NO, IFN-γ, TNF-α, M1 macrophages, protein carbonyl and malondialdehyde tissue levels. The number and size of granulomas in doxycycline-treated animals was higher than untreated and Pzt-treated mice. Exudative/productive granulomas were predominant in untreated and doxycycline-treated animals, while fibrotic/involutive granulomas were more frequent in Pzt-treated mice. The reference treatment with Pzt attenuated all these parameters. CONCLUSION Our findings indicated that doxycycline aggravated lung granulomatous inflammation in a dose-dependent way. Although Th1 effectors are protective against several intracellular pathogens, effective schistosomicidal responses are dependent of the Th2 phenotype. Thus, doxycycline contributes to the worsening of lung granulomatous inflammation by potentiating eosinophils influx and downregulating Th2 effectors, reinforcing lipid and protein oxidative damage in chronic S. mansoni infection.
Collapse
Affiliation(s)
- Matheus Augusto Souza
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Reggiani V. Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Eliziária C. Santos
- School of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Minas Gerais, Brazil
| | - Camila C. Campos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Marcos J. Marques
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Raquel L.M. Souza
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Rômulo D. Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil,Corresponding author. Institute of Biomedical Sciences, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas, 37130-000, Minas Gerais, Brazil. Tel.: +55 31 3299 1300.
| |
Collapse
|
11
|
Hananeh WM, Radhi A, Mukbel RM, Ismail ZB. Effects of parasites coinfection with other pathogens on animal host: A literature review. Vet World 2022; 15:2414-2424. [DOI: 10.14202/vetworld.2022.2414-2424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
A parasite-host relationship is complicated and largely remained poorly understood, especially when mixed infections involving pathogenic bacteria and viruses are present in the same host. It has been found that most parasites are able to manipulate the host's immune responses to evade or overcome its defense systems. Several mechanisms have been postulated that may explain this phenomenon in different animal species. Recent evidence suggests that coinfections involving many parasitic species alter the host's vulnerability to other microorganisms, hinder diagnostic accuracy, and may negatively impact vaccination by altering the host's immune responsiveness. The objective of this review was to provide a comprehensive summary of the current understanding of how parasites interact with other pathogens in different animal species. A better understanding of this complex relationship will aid in the improvement efforts of disease diagnosis, treatment, and control measures such as novel and effective vaccines and therapeutics for infectious diseases.
Collapse
Affiliation(s)
- Wael M. Hananeh
- Department of Veterinary Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box, 3030, Irbid 22110, Jordan
| | - Asya Radhi
- Department of Veterinary Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box, 3030, Irbid 22110, Jordan
| | - Rami M. Mukbel
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Zuhair Bani Ismail
- Department of Clinical Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| |
Collapse
|
12
|
Vilas-Boas DF, Oliveira RRG, Gonçalves-Santos E, Silva LS, Diniz LF, Mazzeti AL, Brancaglion GA, Carvalho DT, Caldas S, Novaes RD, Caldas IS. 4-nitrobenzoylcoumarin potentiates the antiparasitic, anti-inflammatory and cardioprotective effects of benznidazole in a murine model of acute Trypanosoma cruzi infection. Acta Trop 2022; 228:106314. [PMID: 35038424 DOI: 10.1016/j.actatropica.2022.106314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 11/29/2022]
Abstract
The anti-inflammatory and cardioprotective potential of coumarin metabolites in infectious myocarditis remains overlooked. Thus, the impact of the synthetic 4-nitrobenzoylcoumarin (4NB) alone and combined with benznidazole (Bz) in a murine model of Trypanosoma cruzi-induced acute myocarditis was investigated. Swiss mice infected with T. cruzi were randomized in 8 groups: uninfected, infected untreated or treated with 50 and 100 mg/kg 4NB or Bz alone and combined. Treatments were administered by gavage for 20 days. Cytokines (IL-2, IL-6, IL-10, IL-17, TNFα, and IFN-γ), immunoglobulin reactivity index (total IgG, IgG1, IgG2a and IgG2b), atrial natriuretic peptide (ANP), parasitemia, serum transaminases, heart and liver cellularity were analyzed. T. cruzi infection induced blood parasitism, heart and liver inflammation, upregulated all cytokines, IgG reactivity index, ANP and transaminase levels, determining 43% mortality in untreated mice. Transaminase levels, mean parasitemia, heart inflammation and ANP were reduced in 4NB-treated mice, reaching a 100% survival rate. Total survival (100%) was also obtained in all combinations of Bz and 4NB, which were effective in reducing blood parasitism, transaminases, cytokines and ANP levels, IgG reactivity index, liver and heart interstitial cellularity compared to 50 mg/kg Bz. Our findings indicated that 4NB alone and combined with Bz was well tolerated, showing no evidence of hepatotoxicity. Mainly in combination, these drugs exerted protective effects against T. cruzi-induced acute myocarditis by attenuating blood parasitism, systemic and heart inflammation. Thus, combinations based on 4NB and Bz are potentially relevant to develop new and more effective drug regimens for the treatment of T. cruzi-induced myocarditis.
Collapse
Affiliation(s)
- Diego F Vilas-Boas
- Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, 37130-001 Alfenas, MG, Brazil
| | - Raphaela R G Oliveira
- Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, 37130-001 Alfenas, MG, Brazil
| | - Elda Gonçalves-Santos
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas, 37130-000 Alfenas, MG, Brazil
| | - Luana S Silva
- Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, 37130-001 Alfenas, MG, Brazil
| | - Lívia F Diniz
- Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, 37130-001 Alfenas, MG, Brazil
| | - Ana L Mazzeti
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, RJ, Brazil
| | - Guilherme A Brancaglion
- Departamento de Alimentos e Medicamentos, Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas, 37130-000 Alfenas, MG, Brazil
| | - Diogo T Carvalho
- Departamento de Alimentos e Medicamentos, Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas, 37130-000 Alfenas, MG, Brazil
| | - Sergio Caldas
- Fundação Ezequiel Dias, 30510-010 Belo Horizonte, MG, Brazil
| | - Rômulo D Novaes
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas, 37130-000 Alfenas, MG, Brazil
| | - Ivo S Caldas
- Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, 37130-001 Alfenas, MG, Brazil.
| |
Collapse
|
13
|
Yu S, Sun L, Jiang J, He X, Zhou Q. Common variants in AGR1 genes contributed to the risk and traits of cirrhotic cardiomyopathy in Han Chinese population. Biomark Med 2022; 16:331-340. [PMID: 35234520 DOI: 10.2217/bmm-2021-0744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aimed to determine the association between polymorphisms of the ARG1 gene and the risk and traits of cirrhotic cardiomyopathy (CCM). Methods: A total of 468 CCM and 1012 cirrhosis patients were enrolled, and 12 single-nucleotide polymorphisms (SNPs) in the ARG1 gene were genotyped. Differences in genotype, allele and haplotype frequencies of the SNPs between the CCM and cirrhosis groups were analyzed by chi-square test. Correlations of the genotypes of SNPs and representative traits of liver and heart function were performed using linear regression analysis. Results: SNPs rs2781666 and rs2781667 were associated with the risk of CCM in both dominant and additive inheritance models. The GG genotype frequency of rs2781666 and CC genotype frequency of rs2781667 were lower in the CCM group than in the cirrhosis group. The G-C haplotype frequency of the block consisting of rs2781666 and rs2781667 was higher and the T-T haplotype frequency was lower in CCM patients than in cirrhosis patients. SNP rs2781666 was associated with the alanine transaminase level, and rs2781667 was associated with the ARG1 level and left atrial diameter. Conclusion: SNPs rs2781666 and rs2781667 in the ARG1 gene were associated with susceptibility to and traits of CCM in the Han Chinese population.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Lei Sun
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jue Jiang
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Xin He
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Qi Zhou
- Department of Ultrasound, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| |
Collapse
|
14
|
Chronic rapamycin pretreatment modulates arginase/inducible nitric oxide synthase balance attenuating aging-dependent susceptibility to Trypanosoma cruzi infection and acute myocarditis. Exp Gerontol 2022; 159:111676. [DOI: 10.1016/j.exger.2021.111676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
|
15
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:1342-1352. [DOI: 10.1093/jpp/rgac006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022]
|
16
|
Bernardes MTCP, Agostini SBN, Pereira GR, da Silva LP, da Silva JB, Bruschi ML, Novaes RD, Carvalho FC. Preclinical study of methotrexate-based hydrogels versus surfactant based liquid crystal systems on psoriasis treatment. Eur J Pharm Sci 2021; 165:105956. [PMID: 34314841 DOI: 10.1016/j.ejps.2021.105956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/29/2021] [Accepted: 07/21/2021] [Indexed: 12/11/2022]
Abstract
Psoriasis is an autoimmune, inflammatory and chronic skin disease in which cell growth and proliferation are increased, causing erythema, lesions and skin's peeling. Oral methotrexate (MTX) is the first-choice drug when phototherapy or retinoid treatment are not effective. Topical administration can be advantageous to better orientate the drug's delivery; however, the stratum corneum performs as a barrier for hydrofilic drugs penetration. This study sought to evaluate two different types of vehicles for MTX on the psoriasis treatment - hydrogel and liquid crystal systems (LCs). Lamellar and hexagonal liquid crystalline phases were selected from a ternary phase diagram based on polysorbate 80, isopropyl miristate and water. The hydrogel was based on alkylated carbomer (ACH). Rheological analysis showed ACH was more elastic than lamellar and hexagonal phases. ACH interacted better with pig skin than LCs in bioadhesion assay. Preclinical study revealed the ACH decreased inflammation in mice with induced psoriasis, being as effective as dexamethasone to regulate epidermis thickness, COX-2 and myeloperoxidase activity and TNF-α level, while LCs demonstrated inflammatory effect. Therefore, MTX-loaded hydrogel based platforms are indicated for local treatment of psoriasis and present great potential for further studies.
Collapse
Affiliation(s)
| | | | - Gislaine Ribeiro Pereira
- Faculdade de Farmácia, Departamento de Fármacos e Alimentos, Universidade Federal de Alfenas, Brazil
| | - Laíla Pereira da Silva
- Instituto de Ciências Biomédicas, Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Brazil
| | - Jéssica Bassi da Silva
- Laboratório de Pesquisa e Desenvolvimento de Sistemas de Liberação de Fármacos, Departamento de Farmácia, Universidade Estadual de Maringá, Brazil
| | - Marcos Luciano Bruschi
- Laboratório de Pesquisa e Desenvolvimento de Sistemas de Liberação de Fármacos, Departamento de Farmácia, Universidade Estadual de Maringá, Brazil
| | - Rômulo Dias Novaes
- Instituto de Ciências Biomédicas, Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Brazil
| | - Flávia Chiva Carvalho
- Faculdade de Farmácia, Departamento de Fármacos e Alimentos, Universidade Federal de Alfenas, Brazil.
| |
Collapse
|
17
|
Santos MP, Gonçalves-Santos E, Gonçalves RV, Santos EC, Campos CC, Bastos DSS, Marques MJ, Souza RLM, Novaes RD. Doxycycline aggravates granulomatous inflammation and lung microstructural remodeling induced by Schistosoma mansoni infection. Int Immunopharmacol 2021; 94:107462. [PMID: 33611055 DOI: 10.1016/j.intimp.2021.107462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/20/2022]
Abstract
Although doxycycline exhibits immunomodulatory properties, its effects on pulmonary infection by Schistosoma mansoni remain overlooked. Thus, we investigated the impact of this drug on lung granulomatous inflammation and microstructural remodeling in a murine model of schistosomiasis. Swiss mice were randomized in four groups: (i) uninfected, (ii) infected with S. mansoni and untreated, (iii) infected treated with praziquantel (Pzq; 200 mg/kg), and (iv) infected treated with Dox (50 mg/kg). Pz was administered in a single dose, and Dox for 60 days. S. mansoni induced marked granulomatous lung inflammation, which was associated to cytokines upregulation (IL-2, IL-4, IL-10, IFN-γ, TNF-α, and TGF-β), neutrophils and macrophages recruitment, alveolar collapse, lung fibrosis, and extensive depletion of elastic fibers. These parameters were attenuated by Pzq and aggravated by Dox. Exudative/productive granulomas were predominant in untreated and Dox-treated animals, while fibrotic granulomas were more frequent in Pzq-treated mice. The number and size of granulomas in Dox-treated animals was higher than untreated and Pzq-treated mice. Dox treatment inhibited the increase in MMP-1 and MMP-2 activity but upregulated myeloperoxidase and N-acetylglucosaminidase activity compared to untreated and Pzq-treated animals. Dox and Pzq exerted no effect on elastin depletion and upregulation of elastase activity. Together, our findings indicated that Dox aggravated granulomatous inflammation, accelerating lung microstructural remodeling by downregulating MMP-1 and MMP-2 activity without impair neutrophils and macrophages recruitment or elastase activity. Thus, Dox potentiates inflammatory damage associated with lung fibrosis, elastin depletion and massive alveolar collapse, profoundly subverting lung structure in S. mansoni-infected mice.
Collapse
Affiliation(s)
- Margarida P Santos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Eliziária C Santos
- School of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Minas Gerais, Brazil
| | - Camila C Campos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Daniel S S Bastos
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Marcos J Marques
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Raquel L M Souza
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Mendonça AAS, Gonçalves-Santos E, Souza-Silva TG, González-Lozano KJ, Caldas IS, Gonçalves RV, Diniz LF, Novaes RD. Thioridazine aggravates skeletal myositis, systemic and liver inflammation in Trypanosoma cruzi-infected and benznidazole-treated mice. Int Immunopharmacol 2020; 85:106611. [PMID: 32447223 DOI: 10.1016/j.intimp.2020.106611] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022]
Abstract
While thioridazine (Tio) inhibits the antioxidant defenses of Trypanosoma cruzi, the gold standard antitrypanosomal drug benznidazole (Bz) has potent anti-inflammatory and pro-oxidant properties. The combination of these drugs has never been tested to determine the effect on T. cruzi infection. Thus, we compared the impact of Tio and Bz, administered alone and in combination, on the development of skeletal myositis and liver inflammation in T. cruzi-infected mice. Swiss mice were randomized into six groups: uninfected untreated, infected untreated, treated with Tio (80 mg/kg) alone, Bz (50 or 100 mg/kg) alone, or a combination of Tio and Bz. Infected animals were inoculated with a virulent T. cruzi strain (Y) and treated by gavage for 20 days. Mice untreated or treated with Tio alone developed the most intense parasitemia, highest parasitic load, elevated IL-10, IL-17, IFN-γ, and TNF-α plasma levels, increased N-acetylglucosaminidase and myeloperoxidase activity in the liver and skeletal muscle, as well as severe myositis and liver inflammation (P < 0.05). All parameters were markedly attenuated in animals receiving Bz alone (P < 0.05). However, the co-administration of Tio impaired the response to Bz chemotherapy, causing a decrease in parasitological control (parasitemia and parasite load), skeletal muscle and liver inflammation, and increased microstructural damage, when compared to the group receiving Bz alone (P < 0.05). Altogether, our findings indicated that Tio aggravates systemic inflammation, skeletal myositis and hepatic inflammatory damage in T. cruzi-infected mice. By antagonizing the antiparasitic potential of Bz, Tio limits the anti-inflammatory, myoprotectant and hepatoprotective effects of the reference chemotherapy, aggravating the pathological remodeling of both organs. As the interaction of T. cruzi infection, Bz and Tio is potentially toxic to the liver, inducing inflammation and microvesicular steatosis; this drug combination represents a worrying pharmacological risk factor in Chagas disease.
Collapse
Affiliation(s)
- Andréa A S Mendonça
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Thaiany G Souza-Silva
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Kelly J González-Lozano
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Ivo S Caldas
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-000, Minas Gerais, Brazil
| | - Lívia F Diniz
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, Minas Gerais, Brazil.
| |
Collapse
|
19
|
Mendonça AAS, Gonçalves-Santos E, Souza-Silva TG, González-Lozano KJ, Caldas IS, Gonçalves RV, Diniz LF, Novaes RD. Could phenothiazine-benznidazole combined chemotherapy be effective in controlling heart parasitism and acute infectious myocarditis? Pharmacol Res 2020; 158:104907. [PMID: 32416214 DOI: 10.1016/j.phrs.2020.104907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Phenothiazines inhibit major antioxidant defense mechanisms in trypanosomatids and exhibit potent cytotoxic effects in vitro. However, the relevance of these drugs in the treatment of Trypanosoma cruzi-induced acute myocarditis is poorly explored, especially in combination with reference trypanocidal drugs. Thus, we compared the antiparasitic and cardioprotective potential of thioridazine (TDZ) and benznidazole (Bz) administered in monotherapy and combined in a murine model of T. cruzi-induced acute myocarditis. Female mice were randomized into six groups: (i) uninfected untreated, (ii) infected untreated, or infected treated with (iii) Bz (100 mg/kg), (iv) TDZ (80 mg/kg), (v) Bz (100 mg/kg) + TDZ (80 mg/kg), or (vi) Bz (50 mg/kg) + TDZ (80 mg/kg). Infected animals were inoculated with 2000 T. cruzi trypomastigotes and treated by gavage for 20 days. Animals that received TDZ alone presented the highest levels of parasitemia, parasitic load and anti-T. cruzi immunoglobulin G titers; cardiac upregulation of N-acetyl-β-D-glucosaminidase activity, nitric oxide, malondialdehyde and cytokines (IFN-γ, TNF-α, IL-10 and IL-17); as well as microstructural damage compared to the other groups (p < 0.05). These parameters were reduced in groups receiving Bz monotherapy compared to the other groups (p < 0.05). The combination of TDZ and Bz attenuated the response to treatment, worsening parasitological control, oxidative heart damage and myocarditis compared to the group treated with Bz alone (p < 0.05). Our results indicate that when administered alone, TDZ potentiated the pathological outcomes in animals infected with T. cruzi. Moreover, TDZ attenuated the antiparasitic effect of Bz when administered together, impairing parasitological control, potentiating inflammation, molecular oxidation and pathological microstructural remodeling of the heart. Thus, our findings indicate that TDZ acts as a pharmacological risk factor and Bz-based monotherapy remains a better cardioprotective drug against Trypanosoma cruzi-induced acute myocarditis.
Collapse
Affiliation(s)
- Andréa A S Mendonça
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Thaiany G Souza-Silva
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Kelly J González-Lozano
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Ivo S Caldas
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa, 36570-000 Minas Gerais, Brazil
| | - Lívia F Diniz
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil.
| |
Collapse
|
20
|
Coakley G, Harris NL. Interactions between macrophages and helminths. Parasite Immunol 2020; 42:e12717. [PMID: 32249432 DOI: 10.1111/pim.12717] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Macrophages, the major population of tissue-resident mononuclear phagocytes, contribute significantly to the immune response during helminth infection. Alternatively activated macrophages (AAM) are induced early in the anti-helminth response following tissue insult and parasite recognition, amplifying the early type 2 immune cascade initiated by epithelial cells and ILC2s, and subsequently driving parasite expulsion. AAM also contribute to functional alterations in tissues infiltrated with helminth larvae, mediating both tissue repair and inflammation. Their activation is amplified and occurs more rapidly following reinfection, where they can play a dual role in trapping tissue migratory larvae and preventing or resolving the associated inflammation and damage. In this review, we will address both the known and emerging roles of tissue macrophages during helminth infection, in addition to considering both outstanding research questions and new therapeutic strategies.
Collapse
Affiliation(s)
- Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, The Alfred Centre The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| | - Nicola Laraine Harris
- Department of Immunology and Pathology, Central Clinical School, The Alfred Centre The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
de Rezende MC, Moreira JMP, Fernandes LLM, Rodrigues VF, Negrão-Corrêa D. Strongyloides venezuelensis-infection alters the profile of cytokines and liver inflammation in mice co-infected with Schistosoma mansoni. Cytokine 2020; 127:154931. [DOI: 10.1016/j.cyto.2019.154931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
|
22
|
Dias MV, Castro AP, Campos CC, Souza-Silva TG, Gonçalves RV, Souza RLM, Marques MJ, Novaes RD. Doxycycline hyclate: A schistosomicidal agent in vitro with immunomodulatory potential on granulomatous inflammation in vivo. Int Immunopharmacol 2019; 70:324-337. [DOI: 10.1016/j.intimp.2019.02.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/13/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
|
23
|
Impact of Paracoccidioides brasiliensis Coinfection on the Evolution of Schistosoma mansoni-Induced Granulomatous Liver Injury in Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8319465. [PMID: 31019973 PMCID: PMC6451801 DOI: 10.1155/2019/8319465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/11/2019] [Accepted: 03/03/2019] [Indexed: 01/21/2023]
Abstract
The pathogens Schistosoma mansoni and Paracoccidioides brasiliensis share common geographic areas, determining infectious diseases with high mortality rates worldwide. Histopathological and immunological changes induced by each pathogen are well understood; however, the host responses to S. mansoni and P. brasiliensis coinfection are still unknown. Thus, we investigated liver damage and cytokines production in a murine model acutely and chronically coinfected with these pathogens. Fourty male Swiss mice were infected with S. mansoni and P. brasiliensis alone or coinfected. The animals were euthanized with 50 (acute infection) and 120 (chronic infection) days of infection. All infected animals exhibited liver inflammation. Intense granulomatous inflammation was detected in animals infected with S. mansoni alone and those coinfected. Productive and involutive granulomas were clearly observed in acute and chronic infections, respectively. Granuloma size was reduced in the acute phase and increased in the chronic phase of S. mansoni and P. brasiliensis coinfection, compared with animals infected only with S. mansoni. In the chronic phase of infection, the granulomatous inflammation in coinfected animals was characterized by intense neutrophils accumulation and reduced eosinophils number. IFN-γ, IL-2, IL-4, and IL-5 circulating levels were increased in all infected groups. Coinfected animals presented attenuated IFN-γ and IL-4 production in the acute and chronic infections. Taken together, our findings indicate that coinfected animals exhibited a differential modulation of granulomatous inflammation during the acute and chronic phases of infection, which was potentially associated with a divergent profile of cytokines production and migration of neutrophils and eosinophils in response to S. mansoni and P. brasiliensis antigenic stimulation.
Collapse
|
24
|
Li S, Xu S, Li C, Ran X, Cui G, He M, Miao K, Zhao C, Yan J, Hui R, Zhou N, Wang Y, Jiang J, Zhang J, Wang D. A life support-based comprehensive treatment regimen dramatically lowers the in-hospital mortality of patients with fulminant myocarditis: a multiple center study. SCIENCE CHINA-LIFE SCIENCES 2019; 62:369-380. [PMID: 30850929 DOI: 10.1007/s11427-018-9501-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/22/2019] [Indexed: 12/20/2022]
Abstract
Fulminant myocarditis (FM) has unacceptable high mortality. This study aimed to evaluate the therapeutic efficacy of a life support-based comprehensive treatment regimen (LSBCTR), a completely novel treatment regimen, for FM. A total of 169 FM patients recruited from January 2008 to December 2018 were divided into two groups: patients receiving LSBCTR (81 cases), which includes (i) mechanical life support (positive pressure respiration, intra-aortic balloon pump with or without extracorporeal membrane oxygenation), (ii) immunomodulation therapy using sufficient doses of glucocorticoids and immunoglobulins, and (iii) application of neuraminidase inhibitors, and those receiving conventional treatment (88 cases). The endpoints were in-hospital death and heart-transplantation. Of all the population, 44 patients (26.0%) died in hospitals. In-hospital mortality was 3.7% (3/81) for LSBCTR group and 46.6% (41/88) for traditional treatment (P<0.001). Early application of LSBCTR, mechanical life support, neuraminidase inhibitors, and immunomodulation therapy significantly contributed to reduction in in-hospital mortality. This study describes a novel treatment regimen for FM patients that dramatically reduces in-hospital mortality. Its generalization and clinical application will efficiently save lives although further optimization is needed. This study offers an insight that virus infection induced inflammatory waterfall results in cardiac injury and cardiogenic shock and is the therapeutic target.
Collapse
Affiliation(s)
- Sheng Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Shengyong Xu
- Emergency Department of Union Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Xiao Ran
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Guanglin Cui
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Mengying He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Kun Miao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chunxia Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Rutai Hui
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100037, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Jiangang Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - Jing Zhang
- Fuwai Huazhong Cardiovascular Hospital, Zhengzhou, 451450, China.
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
25
|
Felizardo AA, Caldas IS, Mendonça AAS, Gonçalves RV, Tana FL, Almeida LA, Novaes RD. Impact of Trypanosoma cruzi infection on nitric oxide synthase and arginase expression and activity in young and elderly mice. Free Radic Biol Med 2018; 129:227-236. [PMID: 30248443 DOI: 10.1016/j.freeradbiomed.2018.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/21/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
Elderly organisms are more susceptible to infectious diseases. However, the impact of aging on antiparasitic mechanisms, especially the nitric oxide pathway, is poorly understood. Using an integrated in vivo and in vitro model, we compared the severity of Trypanosoma cruzi infection in young and elderly (8 or 72 weeks old) mice. Forty C57BL/6 mice were randomized into four groups: Y-inf, young infected; Yn-inf, young uninfected; A-inf, aged infected; An-inf, aged uninfected. Parasitemia was measured daily, and animals were euthanized after 15 days of infection. Trypanosoma cruzi-induced inflammatory processes were analyzed in blood and heart samples, as well as in bone marrow-derived macrophages (BMDMs) co-cultured with splenocytes isolated from young or elderly mice. Our results indicated upregulated IgG2b and IL-17 production in elderly animals, which was not sufficient to reduce parasitemia, parasitic load and myocarditis to levels observed in young animals. The higher susceptibility of elderly mice to T. cruzi infection was accompanied by reduced cardiac inducible nitric oxide synthase (iNOS) gene expression, nitric oxide (NO) and IFN-γ levels, as well as an antagonistic upregulation of arginase-1 expression and arginase activity. The same responses were observed when BMDMs co-cultured with splenocytes from elderly mice were stimulated with T. cruzi antigens. Our findings indicate that elderly mice were more susceptible to T. cruzi infection, which was potentially related to an attenuated response to antigenic stimulation, inhibition of iNOS gene expression and NO production, and antagonistic upregulation of arginase gene expression and activity, which created favorable conditions for heart parasitism and myocarditis development.
Collapse
Affiliation(s)
- Amanda A Felizardo
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Ivo S Caldas
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Andréa A S Mendonça
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-000, Minas Gerais, Brazil
| | - Fernanda L Tana
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Leonardo A Almeida
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil.
| |
Collapse
|
26
|
Mabbott NA. The Influence of Parasite Infections on Host Immunity to Co-infection With Other Pathogens. Front Immunol 2018; 9:2579. [PMID: 30467504 PMCID: PMC6237250 DOI: 10.3389/fimmu.2018.02579] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Parasites have evolved a wide range of mechanisms that they use to evade or manipulate the host's immune response and establish infection. The majority of the in vivo studies that have investigated these host-parasite interactions have been undertaken in experimental animals, especially rodents, which were housed and maintained to a high microbiological status. However, in the field situation it is increasingly apparent that pathogen co-infections within the same host are a common occurrence. For example, chronic infection with pathogens including malarial parasites, soil-transmitted helminths, Mycobacterium tuberculosis and viruses such as HIV may affect a third of the human population of some developing countries. Increasing evidence shows that co-infection with these pathogens may alter susceptibility to other important pathogens, and/or influence vaccine efficacy through their effects on host immune responsiveness. Co-infection with certain pathogens may also hinder accurate disease diagnosis. This review summarizes our current understanding of how the host's immune response to infection with different types of parasites can influence susceptibility to infection with other pathogenic microorganisms. A greater understanding of how infectious disease susceptibility and pathogenesis can be influenced by parasite co-infections will enhance disease diagnosis and the design of novel vaccines or therapeutics to more effectively control the spread of infectious diseases.
Collapse
Affiliation(s)
- Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Purinergic Antagonist Suramin Aggravates Myocarditis and Increases Mortality by Enhancing Parasitism, Inflammation, and Reactive Tissue Damage in Trypanosoma cruzi-Infected Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7385639. [PMID: 30364017 PMCID: PMC6186315 DOI: 10.1155/2018/7385639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022]
Abstract
Suramin (Sur) acts as an ecto-NTPDase inhibitor in Trypanosoma cruzi and a P2-purinoceptor antagonist in mammalian cells. Although the potent antitrypanosomal effect of Sur has been shown in vitro, limited evidence in vivo suggests that this drug can be dangerous to T. cruzi-infected hosts. Therefore, we investigated the dose-dependent effect of Sur-based chemotherapy in a murine model of Chagas disease. Seventy uninfected and T. cruzi-infected male C57BL/6 mice were randomized into five groups: SAL = uninfected; INF = infected; SR5, SR10, and SR20 = infected treated with 5, 10, or 20 mg/kg Sur. In addition to its effect on blood and heart parasitism, the impact of Sur-based chemotherapy on leucocytes myocardial infiltration, cytokine levels, antioxidant defenses, reactive tissue damage, and mortality was analyzed. Our results indicated that animals treated with 10 and 20 mg/kg Sur were disproportionally susceptible to T. cruzi, exhibiting increased parasitemia and cardiac parasitism (amastigote nests and parasite load (T. cruzi DNA)), intense protein, lipid and DNA oxidation, marked myocarditis, and mortality. Animals treated with Sur also exhibited reduced levels of nonprotein antioxidants. However, the upregulation of catalase, superoxide dismutase, and glutathione-S-transferase was insufficient to counteract reactive tissue damage and pathological myocardial remodeling. It is still poorly understood whether Sur exerts a negative impact on the purinergic signaling of T. cruzi-infected host cells. However, our findings clearly demonstrated that through enhanced parasitism, inflammation, and reactive tissue damage, Sur-based chemotherapy contributes to aggravating myocarditis and increasing mortality rates in T. cruzi-infected mice, contradicting the supposed relevance attributed to this drug for the treatment of Chagas disease.
Collapse
|
28
|
Mendonça AAS, Coelho CM, Veloso MP, Caldas IS, Gonçalves RV, Teixeira AL, de Miranda AS, Novaes RD. Relevance of Trypanothione Reductase Inhibitors on Trypanosoma cruzi Infection: A Systematic Review, Meta-Analysis, and In Silico Integrated Approach. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8676578. [PMID: 30473742 PMCID: PMC6220389 DOI: 10.1155/2018/8676578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/14/2018] [Indexed: 02/08/2023]
Abstract
Due to the rudimentary antioxidant defenses in Trypanosoma cruzi, disruptors of redox balance are promising candidates for new antitrypanosomal drugs. We developed an integrated model based on systematic review, meta-analyses, and molecular modeling to evaluate the effect of trypanothione reductase (TR) inhibitors in T. cruzi infections. Our findings indicated that the TR inhibitors analyzed were effective in reducing parasitemia and mortality due to Trypanosoma cruzi infection in animal models. The most investigated drugs (clomipramine and thioridazine) showed no beneficial effects on the occurrence of infection-related electrocardiographic abnormalities or the affinity and density of cardiac β-adrenergic receptors. The affinity between the tested ligands and the active site of TR was confirmed by molecular docking. However, the molecular affinity score was unable to explain TR inhibition and T. cruzi death in vitro or the antiparasitic potential of these drugs when tested in preclinical models of T. cruzi infection. The divergence of in silico, in vitro, and in vivo findings indicated that the anti-T. cruzi effects of the analyzed drugs were not restricted to TR inhibition. As in vivo studies on TR inhibitors are still scarce and exhibit methodological limitations, mechanistic and highly controlled studies are required to improve the quality of evidence.
Collapse
Affiliation(s)
- Andréa Aparecida Santos Mendonça
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
- Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Camila Morais Coelho
- Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Marcia Paranho Veloso
- Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Ivo Santana Caldas
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
- Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | | | - Antônio Lucio Teixeira
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Aline Silva de Miranda
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
- Institute of Biological Sciences, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Rômulo Dias Novaes
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
- Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| |
Collapse
|
29
|
Rocha Pereira AE, Rodrigues MÂ, Novaes RD, Caldas IS, Martins Souza RL, Costa Pereira AA. Lipopolysaccharide-induced acute lung injury in mice chronically infected by Schistosoma mansoni. Exp Parasitol 2017; 178:21-29. [PMID: 28533109 DOI: 10.1016/j.exppara.2017.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/04/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023]
Abstract
We used a murine model of Schistosoma mansoni (SM) infection and lipopolysaccharide (LPS)-induced endotoxicity to investigate if these conditions can interact to modify the pathological manifestations typically observed in each condition. Swiss mice were randomized into four groups: SAL, uninfected; SM, infected; LPS, uninfected + LPS; and SM + LPS, infected + LPS. S. mansoni infection developed over 120 days, after which blood samples and lungs were collected, peritoneal leukocytes were isolated and cultivated for 6 and 24 h after LPS inoculation (1 mL/kg). Infected animals presented marked granulomatous inflammation. LPS exposure transiently modified the profile of leucocyte migration into the lung tissue and increased NO production by isolated leukocytes, without inducing any acute effect on the structure of schistosomiasis granulomas. Beyond modifying lung morphology, S. mansoni and LPS interacted to modulate the circulating levels of cytokines. S. mansoni infection restricted INF-γ upregulation 6 and 24 h after LPS administration. Conversely, 24 h after inoculation, LPS increased IL-2 and IL-5 levels. Our findings indicate that LPS impaired the lung microenvironment by acutely disrupting inflammatory homeostatic mechanisms that control lung schistosomiasis. As schistosomiasis develops as a chronic condition, long-term exposure to endotoxins could aggravate the granulomatous process, an issue that requires further investigation.
Collapse
Affiliation(s)
- Amanda Esteves Rocha Pereira
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Minas Gerais, 37130-001, Brazil
| | - Maria Ângela Rodrigues
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Minas Gerais, 37130-001, Brazil
| | - Rômulo Dias Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Minas Gerais, 37130-001, Brazil
| | - Ivo Santana Caldas
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Minas Gerais, 37130-001, Brazil
| | - Raquel Lopes Martins Souza
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Minas Gerais, 37130-001, Brazil
| | - Alessandro Antônio Costa Pereira
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Minas Gerais, 37130-001, Brazil.
| |
Collapse
|