Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-33649109

RESUMEN

Cerebral malaria (CM) is a severe immunovasculopathy which presents high mortality rate (15-20%), despite the availability of artemisinin-based therapy. More effective immunomodulatory and/or antiparasitic therapies are urgently needed. Experimental Cerebral Malaria (ECM) in mice is used to elucidate aspects involved in this pathology since manifests many of the neurological features of CM. In the present study, we evaluated the potential mechanisms involved in the protection afforded by perillyl alcohol (POH) in mouse strains susceptible to CM caused by Plasmodium berghei ANKA (PbA) infection through intranasal preventive treatment. Additionally, to evaluate the interaction of POH with the cerebral endothelium using an in vitro model of human brain endothelial cells (HBEC). Pharmacokinetic approaches demonstrated constant and prolonged levels of POH in the plasma and brain after a single intranasal dose. Treatment with POH effectively prevented vascular dysfunction. Furthermore, treatment with POH reduced the endothelial cell permeability and PbA s in the brain and spleen. Finally, POH treatment decreased the accumulation of macrophages and T and B cells in the spleen and downregulated the expression of endothelial adhesion molecules (ICAM-1, VCAM-1, and CD36) in the brain. POH is a potent monoterpene that prevents cerebrovascular dysfunction in vivo and in vitro, decreases parasite sequestration, and modulates different processes related to the activation, permeability, and integrity of the blood brain barrier (BBB), thereby preventing cerebral oedema and inflammatory infiltrates.

3.
FASEB J ; 32(8): 4470-4481, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29558201

RESUMEN

Cerebral malaria (CM) is a multifactorial syndrome involving an exacerbated proinflammatory status, endothelial cell activation, coagulopathy, hypoxia, and accumulation of leukocytes and parasites in the brain microvasculature. Despite significant improvements in malaria control, 15% of mortality is still observed in CM cases, and 25% of survivors develop neurologic sequelae for life-even after appropriate antimalarial therapy. A treatment that ameliorates CM clinical signs, resulting in complete healing, is urgently needed. Previously, we showed a hyperbaric oxygen (HBO)-protective effect against experimental CM. Here, we provide molecular evidence that HBO targets brain endothelial cells by decreasing their activation and inhibits parasite and leukocyte accumulation, thus improving cerebral microcirculatory blood flow. HBO treatment increased the expression of aryl hydrocarbon receptor over hypoxia-inducible factor 1-α (HIF-1α), an oxygen-sensitive cytosolic receptor, along with decreased indoleamine 2,3-dioxygenase 1 expression and kynurenine levels. Moreover, ablation of HIF-1α expression in endothelial cells in mice conferred protection against CM and improved survival. We propose that HBO should be pursued as an adjunctive therapy in CM patients to prolong survival and diminish deleterious proinflammatory reaction. Furthermore, our data support the use of HBO in therapeutic strategies to improve outcomes of non-CM disorders affecting the brain.-Bastos, M. F., Kayano, A. C. A. V., Silva-Filho, J. L., Dos-Santos, J. C. K., Judice, C., Blanco, Y. C., Shryock, N., Sercundes, M. K., Ortolan, L. S., Francelin, C., Leite, J. A., Oliveira, R., Elias, R. M., Câmara, N. O. S., Lopes, S. C. P., Albrecht, L., Farias, A. S., Vicente, C. P., Werneck, C. C., Giorgio, S., Verinaud, L., Epiphanio, S., Marinho, C. R. F., Lalwani, P., Amino, R., Aliberti, J., Costa, F. T. M. Inhibition of hypoxia-associated response and kynurenine production in response to hyperbaric oxygen as mechanisms involved in protection against experimental cerebral malaria.


Asunto(s)
Encéfalo/metabolismo , Hipoxia/metabolismo , Quinurenina/metabolismo , Malaria Cerebral/metabolismo , Oxígeno/metabolismo , Animales , Circulación Cerebrovascular/fisiología , Células Endoteliales/metabolismo , Femenino , Oxigenoterapia Hiperbárica/métodos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Microcirculación/fisiología
4.
PLoS Pathog ; 12(12): e1006054, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27926944

RESUMEN

Malaria remains one of the greatest burdens to global health, causing nearly 500,000 deaths in 2014. When manifesting in the lungs, severe malaria causes acute lung injury/acute respiratory distress syndrome (ALI/ARDS). We have previously shown that a proportion of DBA/2 mice infected with Plasmodium berghei ANKA (PbA) develop ALI/ARDS and that these mice recapitulate various aspects of the human syndrome, such as pulmonary edema, hemorrhaging, pleural effusion and hypoxemia. Herein, we investigated the role of neutrophils in the pathogenesis of malaria-associated ALI/ARDS. Mice developing ALI/ARDS showed greater neutrophil accumulation in the lungs compared with mice that did not develop pulmonary complications. In addition, mice with ALI/ARDS produced more neutrophil-attracting chemokines, myeloperoxidase and reactive oxygen species. We also observed that the parasites Plasmodium falciparum and PbA induced the formation of neutrophil extracellular traps (NETs) ex vivo, which were associated with inflammation and tissue injury. The depletion of neutrophils, treatment with AMD3100 (a CXCR4 antagonist), Pulmozyme (human recombinant DNase) or Sivelestat (inhibitor of neutrophil elastase) decreased the development of malaria-associated ALI/ARDS and significantly increased mouse survival. This study implicates neutrophils and NETs in the genesis of experimentally induced malaria-associated ALI/ARDS and proposes a new therapeutic approach to improve the prognosis of severe malaria.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Neutrófilos/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Lesión Pulmonar Aguda/microbiología , Animales , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Técnica del Anticuerpo Fluorescente , Malaria/complicaciones , Malaria/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Reacción en Cadena de la Polimerasa , Síndrome de Dificultad Respiratoria/microbiología
5.
Vet Res ; 49(1): 6, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29351812

RESUMEN

Anaplasma marginale (A. marginale) has a remarkable impact on livestock production, and an effective vaccine is not currently available due to the inexistence of a small animal model. Recently, BALB/c mice were successfully infected with A. marginale, resulting in an acute and persistent anaplasmosis infection. Here, we designed a hybrid protein containing repeats of polypeptide 1a from major surface protein-1 complex (MSP1a) repeats and common epitopes of outer membrane proteins (OMPs) OMP7, OMP8 and OMP9 expressed in Escherichia coli. Our proof-of-concept assessed vaccinal effectiveness against a challenge with live bacteria. The MSP1a/OMP7/8/9 immunized BALB/C mice exhibited a strong reduction in rickettsemia and had no signs of anaplasmosis or hepatic lesions. In contrast, the non-immunized mice exhibited signs of anaplasmosis and a body weight loss associated with increases in monocyte and neutrophil counts. Furthermore, the non-immunized mice displayed atrophies with chronic inflammatory infiltrates in the spleen and increased binucleation and hydropic degeneration in the hepatocytes. Our findings demonstrated that immunization with our hybrid protein induced a strong reduction in rickettsemia and conferred protection against anaplasmosis. Therefore, given the strong evidence of the protective effect against anaplasmosis, hybrid protein designs are potential candidates for the rational design of vaccinal subunits.


Asunto(s)
Anaplasmosis/prevención & control , Proteínas de la Membrana Bacteriana Externa/inmunología , Epítopos/inmunología , Secuencia de Aminoácidos , Anaplasma marginale/fisiología , Anaplasmosis/inmunología , Anaplasmosis/microbiología , Animales , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/microbiología , Enfermedades de los Bovinos/prevención & control , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Ratas
6.
Malar J ; 16(1): 253, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28619120

RESUMEN

BACKGROUND: Plasmodium vivax parasites are the predominant cause of malaria infections in the Brazilian Amazon. Infected individuals are treated with primaquine, which can induce haemolytic anaemia in glucose-6-phosphate dehydrogenase (G6PD)-deficient individuals and may lead to severe and fatal complications. This X-linked disorder is distributed globally and is caused by allelic variants with a geographical distribution that closely reflects populations exposed historically to endemic malaria. In Brazil, few studies have reported the frequency of G6PD deficiency (G6PDd) present in malaria-endemic areas. This is particularly important, as G6PDd screening is not currently performed before primaquine treatment. The aim of this study was to determine the prevalence of G6PDd in the region of Alto do Juruá, in the Western Brazilian Amazon, an area characterized by a high prevalence of P. vivax infection. METHODS: Five-hundred and sixteen male volunteers were screened for G6PDd using the fluorescence spot test (Beutler test) and CareStart™ G6PD Biosensor system. Demographic and clinical-epidemiological data were acquired through an individual interview. To assess the genetic basis of G6PDd, 24 SNPs were genotyped using the Kompetitive Allele Specific PCR assay. RESULTS: Twenty-three (4.5%) individuals were G6PDd. No association was found between G6PDd and the number of malaria cases. An increased risk of reported haemolysis symptoms and blood transfusions was evident among the G6PDd individuals. Twenty-two individuals had the G6PDd A(-) variant and one the G6PD A(+) variant. The Mediterranean variant was not present. Apart from one polymorphism, almost all SNPs were monomorphic or with low frequencies (0-0.04%). No differences were detected among ethnic groups. CONCLUSIONS: The data indicates that ~1/23 males from the Alto do Juruá could be G6PD deficient and at risk of haemolytic anaemia if treated with primaquine. G6PD A(-) is the most frequent deficiency allele in this population. These results concur with reported G6PDd in other regions in Brazil. Routine G6PDd screening to personalize primaquine administration should be considered, particularly as complete treatment of patients with vivax malaria using chloroquine and primaquine, is crucial for malaria elimination.


Asunto(s)
Deficiencia de Glucosafosfato Deshidrogenasa/genética , Malaria Vivax/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Anemia Hemolítica/inducido químicamente , Antimaláricos/efectos adversos , Antimaláricos/uso terapéutico , Brasil/epidemiología , Estudios Transversales , Enfermedades Endémicas , Genotipo , Deficiencia de Glucosafosfato Deshidrogenasa/epidemiología , Humanos , Malaria Vivax/tratamiento farmacológico , Malaria Vivax/epidemiología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Prevalencia , Primaquina/efectos adversos , Primaquina/uso terapéutico , Adulto Joven
7.
Parasitology ; 144(5): 698-705, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28073384

RESUMEN

Magellanic penguins (Spheniscus magellanicus) are native to Argentina, Chile and the Falkland Islands. Magellanic penguins are highly susceptible to blood parasites such as the mosquito-borne Plasmodium spp., which have been documented causing high morbidity and mortality in zoos and rehabilitation centres. However, to date no blood parasites have been detected in wild Magellanic penguins, and it is not clear whether this is reflective of their true absence or is instead related to an insufficiency in sampling effort or a failure of the diagnostic methods. We examined blood smears of 284 Magellanic penguins from the Argentinean coast and tested their blood samples with nested polymerase chain reaction tests targeting Haemoproteus, Plasmodium, Leucocytozoon and Babesia. No blood parasites were detected. Analysing the sampling effort of previous studies and the climatogeography of the region, we found there is strong basis to conclude that haemosporidians do not infect wild Magellanic penguins on the Argentinean coast. However, at present it is not possible to determine whether such parasites occur on the Chilean coast and at the Falkland Islands. Furthermore, it is troubling that the northward distribution expansion of Magellanic penguins and the poleward distribution shift of vectors may lead to novel opportunities for the transmission of blood parasites.


Asunto(s)
Apicomplexa/aislamiento & purificación , Enfermedades de las Aves/parasitología , Culicidae/parasitología , Insectos Vectores/parasitología , Infecciones Protozoarias en Animales/parasitología , Spheniscidae/parasitología , Animales , Argentina , Babesia/aislamiento & purificación , Clima , Geografía , Haemosporida/aislamiento & purificación , Parasitemia/veterinaria , Plasmodium/aislamiento & purificación , Estudios Prospectivos
8.
Malar J ; 15: 93, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26883507

RESUMEN

BACKGROUND: Communicating the presence of potential zoonotic pathogens such as Plasmodium spp. in wild animals is important for developing both animal and human health policies. METHODS: The translocation of an exotic and invasive population of Leontopithecus chrysomelas (golden-headed lion tamarins) required the screening of these animals for specific pathogens. This studies objective was to investigate Plasmodium spp. infection in the L. chrysomelas, both to know its prevalence in these animals in the local area and to minimize the risk of pathogens being translocated to the destination site. To investigate Plasmodium spp. infection, blood samples from 268 animals were assessed for the presence of Plasmodium spp. by genus-specific PCR and stained thick and thin blood smears were examined by light microscopy. Data of human malaria infection in the studied region was also assembled from SINAN (Diseases Information System Notification-Ministry of Health of Brazil). RESULTS: Results from the PCR and microscopy were all negative and suggested that no L. chrysomelas was infected with Plasmodium spp. Analysis of SINAN data showed that malaria transmission is present among the human population in the studied region. CONCLUSIONS: This study is the first to provide information on Plasmodium spp. infection in L. chrysomelas. Plasmodium spp. infection of this species is rare or absent though malaria parasites circulate in the region. In addition, there is minimal risk of translocating Plasmodium spp. infected animals to the destination site.


Asunto(s)
Bosques , Leontopithecus/parasitología , Malaria/epidemiología , Enfermedades de los Monos/epidemiología , Plasmodium/fisiología , Zoonosis/prevención & control , Animales , Brasil , Malaria/prevención & control , Malaria/transmisión , Enfermedades de los Monos/prevención & control , Enfermedades de los Monos/transmisión , Prevalencia
9.
Mediators Inflamm ; 2016: 4158698, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27974865

RESUMEN

Malaria is a serious disease, caused by the parasite of the genus Plasmodium, which was responsible for 440,000 deaths in 2015. Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is one of the main clinical complications in severe malaria. The murine model DBA/2 reproduces the clinical signs of ALI/ARDS in humans, when infected with Plasmodium berghei ANKA. High levels of HO-1 were reported in cases of severe malaria. Our data indicated that the HO-1 mRNA and protein expression are increased in mice that develop malaria-associated ALI/ARDS (MA-ALI/ARDS). Additionally, the hemin, a HO-1 inducing drug, prevented mice from developing MA-ALI/ARDS when administered prior to the development of MA-ALI/ARDS in this model. Also, hemin treatment showed an amelioration of respiratory parameters in mice, high VEGF levels in the sera, and a decrease in vascular permeability in the lung, which are signs of ALI/ARDS. Therefore, the induction of HO-1 before the development of MA-ALI/ARDS could be protective. However, the increased expression of HO-1 on the onset of MA-ALI/ARDS development may represent an effort to revert the phenotype of this syndrome by the host. We therefore confirm that HO-1 inducing drugs could be used for prevention of MA-ALI/ARDS in humans.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Hemo-Oxigenasa 1/metabolismo , Pulmón/metabolismo , Malaria/metabolismo , Proteínas de la Membrana/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Lesión Pulmonar Aguda/complicaciones , Animales , Permeabilidad Capilar , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hemina/metabolismo , Pulmón/irrigación sanguínea , Malaria/complicaciones , Masculino , Ratones , Ratones Endogámicos DBA , Permeabilidad , Fenotipo , Plasmodium berghei , Síndrome de Dificultad Respiratoria/complicaciones
10.
Vet Res ; 46: 30, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25888987

RESUMEN

Seabird rehabilitation is a valuable strategy to mitigate the impacts of oil pollution and other anthropogenic factors, and can significantly contribute to the conservation of penguins. However, infectious diseases such as avian malaria (Plasmodium spp.) can hamper the success of rehabilitation efforts. We combined morphological and molecular diagnostic methods to investigate the epidemiology and pathology of Plasmodium in Magellanic penguins (Spheniscus magellanicus) at rehabilitation centers along 2500 km of the coastline of Brazil. True prevalence of malarial parasites was estimated between 6.6% and 13.5%. We identified five species, three of which had not been described infecting penguins (P. cathemerium, P. nucleophilum, P. unalis); an additional five distinct Plasmodium lineages were also distinguished, and albeit unidentified these clearly correspond to species that also have not yet been reported in penguins. Our results indicate that the diversity of plasmodia that may infect these birds is greater than previously recognised. Considering the well-defined seasonality observed in this study, it is clear that rehabilitation centers could benefit by narrowing their preventative efforts on penguins maintained or admitted during the Austral spring-summer, particularly by preventing mosquitoes from coming into contact with penguins.


Asunto(s)
Malaria Aviar/epidemiología , Malaria Aviar/patología , Plasmodium/clasificación , Plasmodium/aislamiento & purificación , Spheniscidae , Animales , Brasil/epidemiología , Citocromos b/genética , Citocromos b/metabolismo , Malaria Aviar/parasitología , Datos de Secuencia Molecular , Filogenia , Plasmodium/genética , Plasmodium/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Estaciones del Año , Análisis de Secuencia de ADN/veterinaria
11.
Infect Immun ; 82(2): 830-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478096

RESUMEN

Malaria is a widespread infectious disease caused by the parasite Plasmodium. During pregnancy, malaria infection leads to a range of complications that can affect both the mother and fetus, including stillbirth, infant mortality, and low birth weight. In this study, we utilized a mouse model of placental malaria (PM) infection to determine the importance of the protein MyD88 in the host immune response to Plasmodium during pregnancy. Initially, we demonstrated that Plasmodium berghei NK65GFP adhered to placental tissue via chondroitin sulfate A and induced PM in mice with a C57BL/6 genetic background. To evaluate the involvement of MyD88 in the pathology of PM, we performed a histopathological analysis of placentas obtained from MyD88(-/-) and wild-type (WT) mice following infection on the 19th gestational day. Our data demonstrated that the detrimental placental alterations observed in the infected mice were correlated with the expression of MyD88. Moreover, in the absence of this protein, production of interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) was significantly reduced in the infected mice. More importantly, in contrast to fetuses from infected WT mice, which exhibited a reduction in body weight, the fetuses from infected MyD88(-/-) mice did not display significant weight loss compared to their noninfected littermates. In addition, we observed a decrement of maternal care associated with malaria infection, which was attenuated in the MyD88-deficient mice. Collectively, the results of this study illustrate the pivotal importance of the MyD88 signaling pathway in the pathogenesis of placental malaria, thus presenting new possibilities for targeting MyD88 in therapeutic interventions.


Asunto(s)
Interacciones Huésped-Patógeno , Malaria/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Placenta/inmunología , Plasmodium berghei/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Femenino , Histocitoquímica , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/parasitología , Embarazo , Complicaciones Infecciosas del Embarazo/parasitología
12.
Malar J ; 13: 230, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24927627

RESUMEN

BACKGROUND: The mechanisms through which infection with Plasmodium spp. result in lung disease are largely unknown. Recently a number of mouse models have been developed to research malaria-associated lung injury but no detailed ultrastructure studies of the disease in its terminal stages in a murine model have yet been published. The goal was to perform an ultrastructural analysis of the lungs of mice that died with malaria-associated acute lung injury/acute respiratory distress syndrome to better determine the relevancy of the murine models and investigate the mechanism of disease. METHODS: DBA/2 mice were infected with Plasmodium berghei strain ANKA. Mice had their lungs removed immediately after death, processed using standard methods and viewed by transmission electron microscopy (TEM). RESULTS: Infected red blood cell:endothelium contact, swollen endothelium with distended cytoplasmic extensions and thickening of endothelium basement membrane were observed. Septa were thick and filled with congested capillaries and leukocytes and the alveolar spaces contained blood cells, oedema and cell debris. CONCLUSION: Results show that the lung ultrastructure of P. berghei ANKA-infected mice has similar features to what has been described in post-mortem TEM studies of lungs from individuals infected with Plasmodium falciparum. These data support the use of murine models to study malaria-associated acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/patología , Pulmón/ultraestructura , Malaria/complicaciones , Síndrome de Dificultad Respiratoria/patología , Animales , Modelos Animales de Enfermedad , Pulmón/parasitología , Masculino , Ratones Endogámicos DBA , Microscopía Electrónica de Transmisión , Plasmodium berghei/crecimiento & desarrollo , Plasmodium falciparum
13.
Nat Med ; 13(6): 703-10, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17496899

RESUMEN

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Asunto(s)
Monóxido de Carbono/fisiología , Hemo-Oxigenasa 1/fisiología , Hemo/metabolismo , Malaria Cerebral/enzimología , Animales , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Plasmodium berghei
14.
Mediators Inflamm ; 2014: 912965, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25104883

RESUMEN

Over the past 20 years, the immune effector mechanisms involved in the control of Trypanosoma cruzi, as well as the receptors participating in parasite recognition by cells of the innate immune system, have been largely described. However, the main questions on the physiopathology of Chagas disease remain unanswered: "Why does the host immune system fail to provide sterile immunity?" and "Why do only a proportion of infected individuals develop chronic pathology?" In this review, we describe the mechanisms proposed to explain the inability of the immune system to eradicate the parasite and the elements that allow the development of chronic heart disease. Moreover, we discuss the possibility that the inability of infected cardiomyocytes to sense intracellular T. cruzi contributes to parasite persistence in the heart and the development of chronic pathology.


Asunto(s)
Enfermedad de Chagas/inmunología , Animales , Enfermedad de Chagas/complicaciones , Cardiopatías/etiología , Cardiopatías/inmunología , Cardiopatías/parasitología , Humanos , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/parasitología , Trypanosoma cruzi/inmunología , Trypanosoma cruzi/patogenicidad
15.
Mediators Inflamm ; 2014: 872464, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25276057

RESUMEN

Malaria-associated acute lung injury/acute respiratory distress syndrome (ALI/ARDS) often results in morbidity and mortality. Murine models to study malaria-associated ALI/ARDS have been described; we still lack a method of distinguishing which mice will develop ALI/ARDS before death. This work aimed to characterize malaria-associated ALI/ARDS in a murine model and to demonstrate the first method to predict whether mice are suffering from ALI/ARDS before death. DBA/2 mice infected with Plasmodium berghei ANKA developing ALI/ARDS or hyperparasitemia (HP) were compared using histopathology, PaO2 measurement, pulmonary X-ray, breathing capacity, lung permeability, and serum vascular endothelial growth factor (VEGF) levels according to either the day of death or the suggested predictive criteria. We proposed a model to predict malaria-associated ALI/ARDS using breathing patterns (enhanced pause and frequency respiration) and parasitemia as predictive criteria from mice whose cause of death was known to retrospectively diagnose the sacrificed mice as likely to die of ALI/ARDS as early as 7 days after infection. Using this method, we showed increased VEGF levels and increased lung permeability in mice predicted to die of ALI/ARDS. This proposed method for accurately identifying mice suffering from ALI/ARDS before death will enable the use of this model to study the pathogenesis of this disease.


Asunto(s)
Malaria/complicaciones , Síndrome de Dificultad Respiratoria/patología , Animales , Temperatura Corporal , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos DBA , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Lancet Reg Health Am ; 18: 100407, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36844021

RESUMEN

Background: Each year, 92 million pregnant women are at risk of contracting malaria during pregnancy, with the underestimation of the mortality and morbidity burden associated with Plasmodium vivax. During pregnancy, P. vivax infection is associated with low birth weight, maternal anaemia, premature delivery, and stillbirth. In the State of Acre (Brazil), high transmission leaves pregnant women at greater risk of contracting malaria and having a greater number of recurrences. The study of genetic diversity and the association of haplotypes with adverse pregnancy effects is of great importance for the control of the disease. Here we investigate the genetic diversity of P. vivax parasites infecting pregnant women across their pregnancies. Methods: P. vivax DNA was extracted from 330 samples from 177 women followed during pregnancy, collected in the State of Acre, Brazil. All samples were negative for Plasmodium falciparum DNA. Sequence data for the Pvmsp1 gene was analysed alongside data from six microsatellite (MS) markers. Allelic frequencies, haplotype frequencies, expected heterozygosity (HE) were calculated. Whole genome sequencing (WGS) was conducted on four samples from pregnant women and phylogenetic analysis performed with other samples from South American regions. Findings: Initially, the pregnant women were stratified into two groups-1 recurrence and 2 or more recurrences-in which no differences were observed in clinical gestational outcomes or in placental histological changes between the two groups. Then we evaluated the parasites genetically. An average of 18.5 distinct alleles were found at each of the MS loci, and the HE calculated for each marker indicates a high genetic diversity occurring within the population. There was a high percentage of polyclonal infections (61.7%, 108/175), and one haplotype (H1) occurred frequently (20%), with only 9 of the haplotypes appearing in more than one patient. Interpretation: Most pregnant women had polyclonal infections that could be the result of relapses and/or re-infections. The high percentage of H1 parasites, along with the low frequency of many other haplotypes are suggestive of a clonal expansion. Phylogenetic analysis shows that P. vivax population within pregnant women clustered with other Brazilian samples in the region. Funding: FAPESP and CNPq - Brazil.

17.
Front Immunol ; 14: 1130662, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37122742

RESUMEN

Introduction: Macrophages are central cells in mediating the inflammatory response. Objective and Methods: We evaluated the effect of high glucose conditions on the inflammatory profile and the autophagy pathway in Bone-Marrow Derived Macrophages (BMDM) from diabetic (D-BMDM) (alloxan: 60mg/kg, i.v.) and non-diabetic (ND-BMDM) C57BL/6 mice. BMDM were cultured in medium with normal glucose (5.5 mM), or high glucose (25 mM) concentration and were primed with Nigericin (20µM) stimulated with LPS (100 ng/mL) at times of 30 minutes; 2; 4; 6 and 24 hours, with the measurement of IL-6, IL-1ß and TNF-α cytokines. Results: We have further identified changes in the secretion of pro-inflammatory cytokines IL-6, IL-1ß and TNF-α, where BMDM showed increased secretion of these cytokines after LPS + Nigericin stimulation. In addition, changes were observed in the autophagy pathway, where the increase in the autophagic protein LC3b and Beclin-1 occurred by macrophages of non-diabetic animals in hyperglycemic medium, without LPS stimulation. D-BMDM showed a reduction on the expression of LC3b and Beclin-1, suggesting an impaired autophagic process in these cells. Conclusion: The results suggest that hyperglycemia alters the inflammatory pathways in macrophages stimulated by LPS, playing an important role in the inflammatory response of diabetic individuals.


Asunto(s)
Interleucina-6 , Factor de Necrosis Tumoral alfa , Ratones , Animales , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Beclina-1/metabolismo , Nigericina/farmacología , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Citocinas/metabolismo , Autofagia , Glucosa/metabolismo
18.
Viruses ; 15(4)2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-37112979

RESUMEN

Since December 2019, the world has been experiencing the COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and we now face the emergence of several variants. We aimed to assess the differences between the wild-type (Wt) (Wuhan) strain and the P.1 (Gamma) and Delta variants using infected K18-hACE2 mice. The clinical manifestations, behavior, virus load, pulmonary capacity, and histopathological alterations were analyzed. The P.1-infected mice showed weight loss and more severe clinical manifestations of COVID-19 than the Wt and Delta-infected mice. The respiratory capacity was reduced in the P.1-infected mice compared to the other groups. Pulmonary histological findings demonstrated that a more aggressive disease was generated by the P.1 and Delta variants compared to the Wt strain of the virus. The quantification of the SARS-CoV-2 viral copies varied greatly among the infected mice although it was higher in P.1-infected mice on the day of death. Our data revealed that K18-hACE2 mice infected with the P.1 variant develop a more severe infectious disease than those infected with the other variants, despite the significant heterogeneity among the mice.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Ratones Transgénicos , Pandemias , SARS-CoV-2/genética , Virulencia
19.
Vaccines (Basel) ; 11(3)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36992253

RESUMEN

Acinetobacter baumannii is a Gram-negative, immobile, aerobic nosocomial opportunistic coccobacillus that causes pneumonia, septicemia, and urinary tract infections in immunosuppressed patients. There are no commercially available alternative antimicrobials, and multi-drug resistance is an urgent concern that requires emergency measures and new therapeutic strategies. This study evaluated a multi-drug-resistant A. baumannii whole-cell vaccine, inactivated and adsorbed on an aluminum hydroxide-chitosan (mAhC) matrix, in an A. baumannii sepsis model in immunosuppressed mice by cyclophosphamide (CY). CY-treated mice were divided into immunized, non-immunized, and adjuvant-inoculated groups. Three vaccine doses were given at 0D, 14D, and 28D, followed by a lethal dose of 4.0 × 108 CFU/mL of A. baumannii. Immunized CY-treated mice underwent a significant humoral response, with the highest IgG levels and a higher survival rate (85%); this differed from the non-immunized CY-treated mice, none of whom survived (p < 0.001), and from the adjuvant group, with 45% survival (p < 0.05). Histological data revealed the evident expansion of white spleen pulp from immunized CY-treated mice, whereas, in non-immunized and adjuvanted CY-treated mice, there was more significant organ tissue damage. Our results confirmed the proof-of-concept of the immune response and vaccine protection in a sepsis model in CY-treated mice, contributing to the advancement of new alternatives for protection against A. baumannii infections.

20.
PLoS Pathog ; 6(5): e1000916, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20502682

RESUMEN

The spectrum of the clinical presentation and severity of malaria infections is broad, ranging from uncomplicated febrile illness to severe forms of disease such as cerebral malaria (CM), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), pregnancy-associated malaria (PAM) or severe anemia (SA). Rodent models that mimic human CM, PAM and SA syndromes have been established. Here, we show that DBA/2 mice infected with P. berghei ANKA constitute a new model for malaria-associated ALI. Up to 60% of the mice showed dyspnea, airway obstruction and hypoxemia and died between days 7 and 12 post-infection. The most common pathological findings were pleural effusion, pulmonary hemorrhage and edema, consistent with increased lung vessel permeability, while the blood-brain barrier was intact. Malaria-associated ALI correlated with high levels of circulating VEGF, produced de novo in the spleen, and its blockage led to protection of mice from this syndrome. In addition, either splenectomization or administration of the anti-inflammatory molecule carbon monoxide led to a significant reduction in the levels of sera VEGF and to protection from ALI. The similarities between the physiopathological lesions described here and the ones occurring in humans, as well as the demonstration that VEGF is a critical host factor in the onset of malaria-associated ALI in mice, not only offers important mechanistic insights into the processes underlying the pathology related with malaria but may also pave the way for interventional studies.


Asunto(s)
Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/parasitología , Malaria/patología , Plasmodium berghei , Factor A de Crecimiento Endotelial Vascular/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Obstrucción de las Vías Aéreas/tratamiento farmacológico , Obstrucción de las Vías Aéreas/parasitología , Obstrucción de las Vías Aéreas/patología , Animales , Antiinflamatorios/farmacología , Monóxido de Carbono/farmacología , Modelos Animales de Enfermedad , Disnea/tratamiento farmacológico , Disnea/parasitología , Disnea/patología , Interacciones Huésped-Parásitos , Hipoxia/tratamiento farmacológico , Hipoxia/parasitología , Hipoxia/patología , Pulmón/irrigación sanguínea , Pulmón/parasitología , Pulmón/patología , Malaria/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Plasmodium chabaudi , Plasmodium yoelii , Circulación Pulmonar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA