Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Evolution ; 78(8): 1513-1514, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-38887989

RESUMEN

The virulence of parasites is expected to reflect an evolutionary tradeoff between increasing proliferation rates that enhance transmission and host mortality which curtails transmission. However, host resource availability may also limit parasites' proliferation rate. To understand the role of resource limitation as a driver of virulence evolution, Pak et al. (2024) use a within-host model of red blood cell (RBC) invasion by Plasmodium chabaudi. They find that within-host resource consumption limits the evolution of the parasite's proliferation rate, as the depletion of RBCs during infection results in intermediate optimal virulence. These results suggest that resource limitation, rather than host mortality, may drive the evolution of virulence.


Asunto(s)
Evolución Biológica , Plasmodium chabaudi , Virulencia , Plasmodium chabaudi/genética , Plasmodium chabaudi/patogenicidad , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Animales , Malaria/parasitología
2.
Evolution ; 78(7): 1287-1301, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38581661

RESUMEN

For parasites, robust proliferation within hosts is crucial for establishing the infection and creating opportunities for onward transmission. While faster proliferation enhances transmission rates, it is often assumed to curtail transmission duration by killing the host (virulence), a trade-off constraining parasite evolution. Yet in many diseases, including malaria, the preponderance of infections with mild or absent symptoms suggests that host mortality is not a sufficient constraint, raising the question of what restrains evolution toward faster proliferation. In malaria infections, the maximum rate of proliferation is determined by the burst size, the number of daughter parasites produced per infected red blood cell. Larger burst sizes should expand the pool of infected red blood cells that can be used to produce the specialized transmission forms needed to infect mosquitoes. We use a within-host model parameterized for rodent malaria parasites (Plasmodium chabaudi) to project the transmission consequences of burst size, focusing on initial acute infection where resource limitation and risk of host mortality are greatest. We find that resource limitation restricts evolution toward higher burst sizes below the level predicted by host mortality alone. Our results suggest resource limitation could represent a more general constraint than virulence-transmission trade-offs, preventing evolution towards faster proliferation.


Asunto(s)
Malaria , Plasmodium chabaudi , Animales , Virulencia , Plasmodium chabaudi/genética , Plasmodium chabaudi/patogenicidad , Plasmodium chabaudi/fisiología , Malaria/transmisión , Malaria/parasitología , Malaria/prevención & control , Interacciones Huésped-Parásitos , Evolución Biológica , Eritrocitos/parasitología , Modelos Biológicos
3.
Elife ; 102021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34636723

RESUMEN

It remains challenging to understand why some hosts suffer severe illnesses, while others are unscathed by the same infection. We fitted a mathematical model to longitudinal measurements of parasite and red blood cell density in murine hosts from diverse genetic backgrounds to identify aspects of within-host interactions that explain variation in host resilience and survival during acute malaria infection. Among eight mouse strains that collectively span 90% of the common genetic diversity of laboratory mice, we found that high host mortality was associated with either weak parasite clearance, or a strong, yet imprecise response that inadvertently removes uninfected cells in excess. Subsequent cross-sectional cytokine assays revealed that the two distinct functional mechanisms of poor survival were underpinned by low expression of either pro- or anti-inflammatory cytokines, respectively. By combining mathematical modelling and molecular immunology assays, our study uncovered proximate mechanisms of diverse infection outcomes across multiple host strains and biological scales.


Asunto(s)
Eritrocitos/parasitología , Malaria/parasitología , Plasmodium chabaudi/patogenicidad , Animales , Simulación por Computador , Citocinas/sangre , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Interacciones Huésped-Parásitos , Mediadores de Inflamación/sangre , Malaria/sangre , Malaria/genética , Malaria/inmunología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Modelos Inmunológicos , Carga de Parásitos , Plasmodium chabaudi/inmunología , Índice de Severidad de la Enfermedad , Especificidad de la Especie , Factores de Tiempo
4.
mBio ; 12(5): e0242421, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34607466

RESUMEN

Infections disrupt host metabolism, but the factors that dictate the nature and magnitude of metabolic change are incompletely characterized. To determine how host metabolism changes in relation to disease severity in murine malaria, we performed plasma metabolomics on eight Plasmodium chabaudi-infected mouse strains with diverse disease phenotypes. We identified plasma metabolic biomarkers for both the nature and severity of different malarial pathologies. A subset of metabolic changes, including plasma arginine depletion, match the plasma metabolomes of human malaria patients, suggesting new connections between pathology and metabolism in human malaria. In our malarial mice, liver damage, which releases hepatic arginase-1 (Arg1) into circulation, correlated with plasma arginine depletion. We confirmed that hepatic Arg1 was the primary source of increased plasma arginase activity in our model, which motivates further investigation of liver damage in human malaria patients. More broadly, our approach shows how leveraging phenotypic diversity can identify and validate relationships between metabolism and the pathophysiology of infectious disease. IMPORTANCE Malaria is a severe and sometimes fatal infectious disease endemic to tropical and subtropical regions. Effective vaccines against malaria-causing Plasmodium parasites remain elusive, and malaria treatments often fail to prevent severe disease. Small molecules that target host metabolism have recently emerged as candidates for therapeutics in malaria and other diseases. However, our limited understanding of how metabolites affect pathophysiology limits our ability to develop new metabolite therapies. By providing a rich data set of metabolite-pathology correlations and by validating one of those correlations, our work is an important step toward harnessing metabolism to mitigate disease. Specifically, we showed that liver damage in P. chabaudi-infected mice releases hepatic arginase-1 into circulation, where it may deplete plasma arginine, a candidate malaria therapeutic that mitigates vascular stress. Our data suggest that liver damage may confound efforts to increase levels of arginine in human malaria patients.


Asunto(s)
Arginasa/sangre , Arginasa/metabolismo , Hígado/enzimología , Malaria/sangre , Metabolómica , Plasmodium chabaudi/patogenicidad , Animales , Arginasa/genética , Arginina/metabolismo , Estudios Transversales , Femenino , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL
5.
PLoS Comput Biol ; 16(10): e1008211, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33031367

RESUMEN

To understand why some hosts get sicker than others from the same type of infection, it is essential to explain how key processes, such as host responses to infection and parasite growth, are influenced by various biotic and abiotic factors. In many disease systems, the initial infection dose impacts host morbidity and mortality. To explore drivers of dose-dependence and individual variation in infection outcomes, we devised a mathematical model of malaria infection that allowed host and parasite traits to be linear functions (reaction norms) of the initial dose. We fitted the model, using a hierarchical Bayesian approach, to experimental time-series data of acute Plasmodium chabaudi infection across doses spanning seven orders of magnitude. We found evidence for both dose-dependent facilitation and debilitation of host responses. Most importantly, increasing dose reduced the strength of activation of indiscriminate host clearance of red blood cells while increasing the half-life of that response, leading to the maximal response at an intermediate dose. We also explored the causes of diverse infection outcomes across replicate mice receiving the same dose. Besides random noise in the injected dose, we found variation in peak parasite load was due to unobserved individual variation in host responses to clear infected cells. Individual variation in anaemia was likely driven by random variation in parasite burst size, which is linked to the rate of host cells lost to malaria infection. General host vigour in the absence of infection was also correlated with host health during malaria infection. Our work demonstrates that the reaction norm approach provides a useful quantitative framework for examining the impact of a continuous external factor on within-host infection processes.


Asunto(s)
Interacciones Huésped-Parásitos , Malaria , Anemia/complicaciones , Animales , Teorema de Bayes , Biología Computacional , Femenino , Malaria/complicaciones , Malaria/inmunología , Malaria/parasitología , Malaria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Carga de Parásitos , Plasmodium chabaudi/patogenicidad , Plasmodium chabaudi/fisiología
6.
PLoS Pathog ; 16(4): e1008527, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32348365

RESUMEN

The co-stimulatory molecule ICOS is associated with the induction and regulation of T helper cell responses, including the differentiation of follicular helper T (Tfh) cells and the formation and maintenance of memory T cells. However, the role of ICOS signaling in secondary immune responses is largely unexplored. Here we show that memory T cell formation and maintenance are influenced by persistent infection with P. chabaudi chabaudi AS infection, as memory T cell numbers decline in wild-type and Icos-/- mice after drug-clearance. Following drug-clearance Icos-/- mice display a relapsing parasitemia that occurs more frequently and with higher peaks compared to wild-type mice after re-challenge. The secondary immune response in Icos-/- mice is characterized by significant impairment in the expansion of effector cells with a Tfh-like phenotype, which is associated with a diminished and delayed parasite-specific Ab response and the absence of germinal centers. Similarly, the administration of an anti-ICOSL antagonizing antibody to wild-type mice before and after reinfection with P. c. chabaudi AS leads to an early defect in Tfh cell expansion and parasite-specific antibody production, confirming a need for ICOS-ICOSL interactions to promote memory B cell responses. Furthermore, adoptive transfer of central memory T (TCM) cells from wild-type and Icos-/- mice into tcrb-/- mice to directly evaluate the ability of TCM cells to give rise to Tfh cells revealed that TCM cells from wild-type mice acquire a mixed Th1- and Tfh-like phenotype after P. c. chabaudi AS infection. While TCM cells from Icos-/- mice expand and display markers of activation to a similar degree as their WT counterparts, they displayed a reduced capacity to upregulate markers indicative of a Tfh cell phenotype, resulting in a diminished humoral response. Together these findings verify that ICOS signaling in memory T cells plays an integral role in promoting T cell effector responses during secondary infection with P. c. chabaudi AS.


Asunto(s)
Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Inmunidad Humoral/inmunología , Memoria Inmunológica , Activación de Linfocitos/inmunología , Malaria/inmunología , Malaria/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium chabaudi/metabolismo , Plasmodium chabaudi/patogenicidad , Transducción de Señal , Linfocitos T Colaboradores-Inductores/metabolismo
7.
Proc Natl Acad Sci U S A ; 116(44): 22386-22392, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31615885

RESUMEN

Hosts defend themselves against pathogens by mounting an immune response. Fully understanding the immune response as a driver of host disease and pathogen evolution requires a quantitative account of its impact on parasite population dynamics. Here, we use a data-driven modeling approach to quantify the birth and death processes underlying the dynamics of infections of the rodent malaria parasite, Plasmodium chabaudi, and the red blood cells (RBCs) it targets. We decompose the immune response into 3 components, each with a distinct effect on parasite and RBC vital rates, and quantify the relative contribution of each component to host disease and parasite density. Our analysis suggests that these components are deployed in a coordinated fashion to realize distinct resource-directed defense strategies that complement the killing of parasitized cells. Early in the infection, the host deploys a strategy reminiscent of siege and scorched-earth tactics, in which it both destroys RBCs and restricts their supply. Late in the infection, a "juvenilization" strategy, in which turnover of RBCs is accelerated, allows the host to recover from anemia while holding parasite proliferation at bay. By quantifying the impact of immunity on both parasite fitness and host disease, we reveal that phenomena often interpreted as immunopathology may in fact be beneficial to the host. Finally, we show that, across mice, the components of the host response are consistently related to each other, even when infections take qualitatively different trajectories. This suggests the existence of simple rules that govern the immune system's deployment.


Asunto(s)
Interacciones Huésped-Parásitos/inmunología , Malaria/inmunología , Plasmodium chabaudi/patogenicidad , Reticulocitos/parasitología , Animales , Longevidad , Merozoítos/fisiología , Ratones , Modelos Teóricos , Plasmodium chabaudi/inmunología , Reticulocitos/inmunología
8.
PLoS Pathog ; 15(2): e1007599, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811498

RESUMEN

Plasmodium parasites invade and multiply inside red blood cells (RBC). Through a cycle of maturation, asexual replication, rupture and release of multiple infective merozoites, parasitised RBC (pRBC) can reach very high numbers in vivo, a process that correlates with disease severity in humans and experimental animals. Thus, controlling pRBC numbers can prevent or ameliorate malaria. In endemic regions, circulating parasite-specific antibodies associate with immunity to high parasitemia. Although in vitro assays reveal that protective antibodies could control pRBC via multiple mechanisms, in vivo assessment of antibody function remains challenging. Here, we employed two mouse models of antibody-mediated immunity to malaria, P. yoelii 17XNL and P. chabaudi chabaudi AS infection, to study infection-induced, parasite-specific antibody function in vivo. By tracking a single generation of pRBC, we tested the hypothesis that parasite-specific antibodies accelerate pRBC clearance. Though strongly protective against homologous re-challenge, parasite-specific IgG did not alter the rate of pRBC clearance, even in the presence of ongoing, systemic inflammation. Instead, antibodies prevented parasites progressing from one generation of RBC to the next. In vivo depletion studies using clodronate liposomes or cobra venom factor, suggested that optimal antibody function required splenic macrophages and dendritic cells, but not complement C3/C5-mediated killing. Finally, parasite-specific IgG bound poorly to the surface of pRBC, yet strongly to structures likely exposed by the rupture of mature schizonts. Thus, in our models of humoral immunity to malaria, infection-induced antibodies did not accelerate pRBC clearance, and instead co-operated with splenic phagocytes to block subsequent generations of pRBC.


Asunto(s)
Malaria/inmunología , Malaria/metabolismo , Plasmodium/crecimiento & desarrollo , Animales , Anticuerpos Antiprotozoarios/metabolismo , Modelos Animales de Enfermedad , Eritrocitos/microbiología , Eritrocitos/fisiología , Humanos , Ratones , Parásitos , Fagocitos , Plasmodium/metabolismo , Plasmodium/patogenicidad , Plasmodium chabaudi/inmunología , Plasmodium chabaudi/patogenicidad , Plasmodium yoelii/inmunología , Plasmodium yoelii/patogenicidad
9.
PLoS One ; 13(8): e0202522, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30148845

RESUMEN

Protective immunity to blood-stage malaria is attributed to Plasmodium-specific IgG and effector-memory T helper 1 (Th1) cells. However, mice lacking the costimulatory receptor CD28 (CD28KO) maintain chronic parasitemia at low levels and do not succumb to infection, suggesting that other immune responses contribute to parasite control. We report here that CD28KO mice develop long-lasting non-sterile immunity and survive lethal parasite challenge. This protection correlated with a progressive increase of anti-parasite IgM serum levels during chronic infection. Serum IgM from chronically infected CD28KO mice recognize erythrocytes infected with mature parasites, and effectively control Plasmodium infection by promoting parasite lysis and uptake. These antibodies also recognize autoantigens and antigens from other pathogens. Chronically infected CD28KO mice have high numbers of IgM+ plasmocytes and experienced B cells, exhibiting a germinal-center independent Fas+GL7-CD38+CD73- phenotype. These cells are also present in chronically infected C57BL/6 mice although in lower numbers. Finally, IgM+ experienced B cells from cured C57BL/6 and CD28KO mice proliferate and produce anti-parasite IgM in response to infected erythrocytes. This study demonstrates that CD28 deficiency results in the generation of germinal-center independent IgM+ experienced B cells and the production of protective IgM during experimental malaria, providing evidence for an additional mechanism by which the immune system controls Plasmodium infection.


Asunto(s)
Antígenos CD28/genética , Inmunoglobulina M/inmunología , Malaria/genética , Plasmodium chabaudi/inmunología , 5'-Nucleotidasa/genética , ADP-Ribosil Ciclasa 1/genética , Animales , Anticuerpos Antiprotozoarios/genética , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Diferenciación/genética , Linfocitos B/inmunología , Linfocitos B/parasitología , Antígenos CD28/deficiencia , Antígenos CD28/inmunología , Linfocitos T CD4-Positivos/inmunología , Eritrocitos/parasitología , Centro Germinal/inmunología , Centro Germinal/parasitología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Malaria/sangre , Malaria/inmunología , Malaria/parasitología , Ratones , Ratones Noqueados , Plasmodium chabaudi/patogenicidad , Receptor fas/genética
10.
Cell Host Microbe ; 23(6): 796-808.e6, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29805094

RESUMEN

The Plasmodium cell cycle, wherein millions of parasites differentiate and proliferate, occurs in synchrony with the vertebrate host's circadian cycle. The underlying mechanisms are unknown. Here we addressed this question in a mouse model of Plasmodium chabaudi infection. Inflammatory gene expression and carbohydrate metabolism are both enhanced in interferon-γ (IFNγ)-primed leukocytes and liver cells from P. chabaudi-infected mice. Tumor necrosis factor α (TNFα) expression oscillates across the host circadian cycle, and increased TNFα correlates with hypoglycemia and a higher frequency of non-replicative ring forms of trophozoites. Conversely, parasites proliferate and acquire biomass during food intake by the host. Importantly, cyclic hypoglycemia is attenuated and synchronization of P. chabaudi stages is disrupted in IFNγ-/-, TNF receptor-/-, or diabetic mice. Hence, the daily rhythm of systemic TNFα production and host food intake set the pace for Plasmodium synchronization with the host's circadian cycle. This mechanism indicates that Plasmodium parasites take advantage of the host's feeding habits.


Asunto(s)
Ritmo Circadiano/fisiología , Regulación de la Expresión Génica , Malaria/metabolismo , Plasmodium chabaudi/parasitología , Plasmodium/fisiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/genética , Ciclo Celular/inmunología , Ritmo Circadiano/inmunología , Diabetes Mellitus Experimental , Modelos Animales de Enfermedad , Ingestión de Alimentos , Metabolismo Energético , Glucosa/metabolismo , Interacciones Huésped-Parásitos/inmunología , Interacciones Huésped-Parásitos/fisiología , Hipoglucemia , Insulina/metabolismo , Interferón gamma/metabolismo , Leucocitos/metabolismo , Leucocitos/parasitología , Hígado/metabolismo , Hígado/parasitología , Malaria/inmunología , Ratones , Plasmodium/patogenicidad , Plasmodium chabaudi/patogenicidad , Receptores del Factor de Necrosis Tumoral , Trofozoítos/fisiología
11.
Microb Pathog ; 119: 170-182, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29630927

RESUMEN

Malaria is still a major health problem worldwide. This study aimed to investigate the hepatoprotective role of Indigofera oblongifolia leaf extracts (ILE) against mice hepatic injury induced by Plasmodium chabaudi. Female C57BL/6 mice were treated with 100 mg/kg of ILE after infection with erythrocytes parasitized by P. chabaudi. On day 7 post-infection, the extract improved the histological alteration induced by the parasite. This was evidenced by the decreased histological index induced by ILE. Moreover, ILE was able to increase the hepatic antioxidant capacity and could significantly improve the decrease in erythrocyte count and hemoglobin content in mice blood plasma due to infection. ILE was also able to upregulate the expression of 24 genes related to metabolism and of 3 genes related to the immune response. Furthermore, the extract was able to downregulate the expression of 35 genes related to metabolism and of 82 genes related to immune response. Moreover, the microarray study showed that ILE regulated the change in gene expression induced by the parasite. Among these genes, we quantified the expression of cd209f, cyp7a1, Hsd3b5, Sult2a3, Lcn2, CcI8, Nos2, and saa3-mRNAs. These genes were regulated by ILE. Therefore, our results revealed the protective role of Indigofera oblongifolia against hepatic injury induced by blood stage malaria.


Asunto(s)
Indigofera/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Extractos Vegetales/farmacología , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hígado/lesiones , Hígado/patología , Malaria/parasitología , Malaria/patología , Ratones , Ratones Endogámicos C57BL , Parasitemia , Extractos Vegetales/química , Hojas de la Planta/química , Plasmodium chabaudi/patogenicidad , ARN Mensajero/metabolismo , Regulación hacia Arriba
12.
Drug Metab Dispos ; 46(5): 503-513, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29514828

RESUMEN

This article is a report on a symposium entitled "Physiological Regulation of Drug Metabolism and Transport" sponsored by the American Society for Pharmacology and Experimental Therapeutics and held at the Experimental Biology 2017 meeting in Chicago, IL. The contributions of physiologic and pathophysiological regulation of drug-metabolizing enzymes and transporters to interindividual variability in drug metabolism are increasingly recognized but in many cases are not well understood. The presentations herein discuss the phenomenology, consequences, and mechanism of such regulation. CYP2D6 transgenic mice were used to provide insights into the mechanism of regulation of this enzyme in pregnancy, via hepatocyte nuclear factor 4α, small heterodimer partner, and retinoids. Regulation of intestinal and hepatic drug-processing enzymes by the intestinal microbiota via tryptophan and its metabolites was investigated. The potential impact of parasitic infections on human drug metabolism and clearance was assessed in mice infected with Schistosoma mansoni or Plasmodium chabaudi chabaudi AS, both of which produced widespread and profound effects on murine hepatic drug-metabolizing enzymes. Finally, the induction of Abcc drug efflux transporters by fasting was investigated. This was demonstrated to occur via a cAMP, protein kinase A/nuclear factor-E2-related factor 2/Sirtuin 1 pathway via antioxidant response elements on the Abcc genes.


Asunto(s)
Transporte Biológico/fisiología , Ayuno/fisiología , Inactivación Metabólica/fisiología , Inflamación/fisiopatología , Microbiota/fisiología , Animales , Elementos de Respuesta Antioxidante/fisiología , Citocromo P-450 CYP2D6/metabolismo , Ayuno/metabolismo , Femenino , Microbioma Gastrointestinal/fisiología , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Inflamación/metabolismo , Hígado/metabolismo , Malaria/metabolismo , Malaria/fisiopatología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Tasa de Depuración Metabólica/fisiología , Ratones , Ratones Transgénicos , Plasmodium chabaudi/patogenicidad , Embarazo , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/fisiopatología , Triptófano/metabolismo
13.
Microb Pathog ; 117: 299-303, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29496525

RESUMEN

Malaria is a harmful disease affecting both tropical and subtropical countries and causing sometimes fatal complications. The effects of malaria-related complications on the intestine have been relatively neglected, and the reasons for the intestinal damage caused by malaria infection are not yet clear. The present study aims to evaluate the influence of intestinal vitamin D receptor on host-pathogen interactions during malaria induced in mice by Plasmodium chabaudi. To induce the infection, animals were infected with 106P. chabaudi-parasitized erythrocytes. Mice were sacrificed on day 8 post-infection. The infected mice experienced a significant body weight loss and parasitaemia affecting about 46% of RBCs. Infection caused marked pathological changes in the intestinal tissue indicated by shortening of the intestine and villi. Moreover, the phagocytic activity of macrophages increased significantly (P < 0.01) in the infected villi compared to the non-infected ones. Infection by the parasite also induced marked upregulation of nuclear factor-kappa B, inducible nitric oxide synthase, Vitamin D Receptor, interleukin-1ß, tumour necrosis factor alpha and interferon gamma-mRNA. It can be implied from this that vitamin D receptor has a role in regulating malarial infection.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Mucosa Intestinal/metabolismo , Malaria/sangre , Malaria/complicaciones , Plasmodium chabaudi/patogenicidad , Receptores de Calcitriol/fisiología , Animales , Peso Corporal , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Eritrocitos/patología , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos/genética , Interferón gamma/metabolismo , Interleucina-1beta/metabolismo , Intestinos/parasitología , Intestinos/patología , Macrófagos/metabolismo , Malaria/parasitología , Malaria/patología , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Parasitemia , Fagocitosis , ARN Mensajero/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
14.
Malar J ; 17(1): 18, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29316914

RESUMEN

BACKGROUND: Cerebral malaria and severe anaemia are the most common deadly complications of malaria, and are often associated, both in paediatric and adult patients, with hepatopathy, whose pathogenesis is not well characterized, and sometimes also with acute respiratory distress syndrome (ARDS). Here, two species of murine malaria, the lethal Plasmodium berghei strain NK65 and self-healing Plasmodium chabaudi strain AS which differ in their ability to cause hepatopathy and/or ARDS were used to investigate the lipid alterations, oxidative damage and host immune response during the infection in relation to parasite load and accumulation of parasite products, such as haemozoin. METHODS: Plasma and livers of C57BL/6J mice injected with PbNK65 or PcAS infected erythrocytes were collected at different times and tested for parasitaemia, content of haemozoin and expression of tumour necrosis factor (TNF). Hepatic enzymes, antioxidant defenses and lipids content and composition were also evaluated. RESULTS: In the livers of P. berghei NK65 infected mice both parasites and haemozoin accumulated to a greater extent than in livers of P. chabaudi AS infected mice although in the latter hepatomegaly was more prominent. Hepatic enzymes and TNF were increased in both models. Moreover, in P. berghei NK65 infected mice, increased lipid peroxidation, accumulation of triglycerides, impairment of anti-oxidant enzymes and higher collagen deposition were detected. On the contrary, in P. chabaudi AS infected mice the antioxidant enzymes and the lipid content and composition were normal or even lower than uninfected controls. CONCLUSIONS: This study demonstrates that in C57BL/6J mice, depending on the parasite species, malaria-induced liver pathology results in different manifestations, which may contribute to the different outcomes. In P. berghei NK65 infected mice, which concomitantly develop lethal acute respiratory distress syndrome, the liver tissue is characterized by an excess oxidative stress response and reduced antioxidant defenses while in P. chabaudi AS infected mice hepatopathy does not lead to lipid alterations or reduction of antioxidant enzymes, but rather to inflammation and cytokine burst, as shown earlier, that may favour parasite killing and clearance of the infection. These results may help understanding the different clinical profiles described in human malaria hepatopathy.


Asunto(s)
Hígado/patología , Hígado/parasitología , Malaria/patología , Malaria/parasitología , Plasmodium berghei/patogenicidad , Plasmodium chabaudi/patogenicidad , Animales , Análisis Químico de la Sangre , Enzimas/sangre , Hemoproteínas/análisis , Hígado/enzimología , Pruebas de Función Hepática , Malaria/complicaciones , Ratones Endogámicos C57BL , Síndrome de Dificultad Respiratoria/patología , Factor de Necrosis Tumoral alfa/sangre
15.
Clin Exp Nephrol ; 21(4): 589-596, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27815652

RESUMEN

BACKGROUND: Malaria is an important tropical disease and has remained a serious health problem in many countries. One of the critical complications of malarial infection is renal injury, such as acute renal failure and chronic glomerulopathy. Few animal models of nephropathy related to malarial infection have been reported. Therefore, we developed and investigated a novel malarial nephropathy model in mice infected by murine malaria parasites. METHODS: NC mice and C57BL/6J mice were infected with Ttwo different murine malaria parasites, Plasmodium (P.) chabaudi AS and P. yoelii 17X. After the infection, renal pathology and blood and urinary biochemistry were analyzed. RESULTS: NC mice infected by the murine malaria parasite P. chabaudi AS, but not P. yoelii 17X, developed mesangial proliferative glomerulonephritis with endothelial damage, and decreased serum albumin concentration and increased proteinuria. These pathological changes were accompanied by deposition of immunoglobulin G and complement component 3, mainly in the mesangium until day 4 and in the mesangium and glomerular capillaries from day 8. On day 21, renal pathology developed to focal segmental sclerosis according to light microscopy. In C57BL/6J mice, renal injuries were not observed from either parasite infection. CONCLUSION: The clinical and pathological features of P. chabaudi AS infection in NC mice might be similar to quartan malarial nephropathy resulting from human malaria parasite P. malariae infection. The NC mouse model might therefore be useful in analyzing the underlying mechanisms and developing therapeutic approaches to malaria-related nephropathy.


Asunto(s)
Glomerulonefritis/parasitología , Glomérulos Renales/parasitología , Malaria/parasitología , Plasmodium chabaudi/patogenicidad , Animales , Complemento C3/inmunología , Modelos Animales de Enfermedad , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Interacciones Huésped-Patógeno , Inmunoglobulina G/inmunología , Glomérulos Renales/inmunología , Glomérulos Renales/ultraestructura , Malaria/inmunología , Ratones Endogámicos C57BL , Plasmodium chabaudi/inmunología , Plasmodium chabaudi/ultraestructura , Especificidad de la Especie , Factores de Tiempo
16.
Immun Inflamm Dis ; 4(4): 497-505, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27980783

RESUMEN

INTRODUCTION: Endemic Burkitt's lymphoma (eBL) is associated with Epstein-Barr virus and repeated malaria infections. A defining feature of eBL is the translocation of the c-myc oncogene to the control of the immunoglobulin promoter. Activation-induced cytidine deaminase (AID) has been shown to be critical for this translocation. Malaria infection induces AID in germinal center B cells, but whether malaria infection more broadly affects AID activation in extrafollicular B cells is unknown. METHODS: We either stimulated purified B cells from AID-green fluorescence protein (GFP) reporter mice or infected AID-GFP mice with Plasmodium chabaudi, AID fluorescence was monitored in B cell subsets by flow cytometry. RESULTS: In vitro analysis of B cells from these mice revealed that CpG (a Toll-like receptor 9 ligand) was a potent inducer of AID in both mature and immature B cell subsets. Infection of AID-GFP mice with Plasmodium chabaudi demonstrated that AID expression occurs in transitional and marginal zone B cells during acute malaria infection. Transitional B cells were also capable of differentiating into antibody secreting cells when stimulated in vitro with CpG when isolated from a P. chabaudi-infected mouse. CONCLUSIONS: These data suggest that P. chabaudi is capable of inducing AID expression in B cell subsets that do not participate in the germinal center reaction, suggesting an alternative role for malaria in the etiology of eBL.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Citidina Desaminasa/metabolismo , Plasmodium chabaudi/patogenicidad , Animales , Linfocitos B , Centro Germinal , Malaria , Masculino , Ratones , Ratones Endogámicos C57BL
17.
J Ethnopharmacol ; 193: 517-523, 2016 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-27693771

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Cymbopogon citratus (lemon grass) has been used in traditional medicine as an herbal infusion to treat fever and malaria. Generally, whole plant extracts possess higher biological activity than purified compounds. However, the antimalarial activity of the whole C. citratus plant has not been experimentally tested. AIM OF THE STUDY: To evaluate the antimalarial activity of an herbal infusion and the whole Cymbopogon citratus plant in two experimental models of malaria. MATERIAL AND METHODS: The plant was dried for 10 days at room temperature and was then milled and passed through brass sieves to obtain a powder, which was administered to CBA/Ca mice with a patent Plasmodium chabaudi AS or P. berghei ANKA infection. We analysed the effects of two different doses (1600 and 3200mg/kg) compared with those of the herbal infusion and chloroquine, used as a positive control. We also assessed the prophylactic antimalarial activities of the whole C. citratus plant and the combination of the whole plant and chloroquine. RESULTS: The C. citratus whole plant exhibited prolonged antimalarial activity against both P. chabaudi AS and P. berghei ANKA. The low dose of the whole C. citratus plant displayed higher antimalarial activity than the high dose against P. berghei ANKA. As a prophylactic treatment, the whole plant exhibited higher antimalarial activity than either the herbal infusion or chloroquine. In addition, the combination of the whole C. citratus plant and chloroquine displayed higher activity than chloroquine alone against P. berghei ANKA patent infection. CONCLUSIONS: We demonstrated the antimalarial activity of the whole C. citratus plant in two experimental models. The whole C. citratus plant elicited higher anti-malarial activity than the herbal infusion or chloroquine when used as a prophylactic treatment. The antimalarial activity of the whole C. citratus plant supports continued efforts towards developing whole plant therapies for the management of malaria and other infectious diseases prevalent in resource-poor communities.


Asunto(s)
Antimaláricos/farmacología , Cymbopogon/química , Malaria/tratamiento farmacológico , Extractos Vegetales/farmacología , Plasmodium berghei/efectos de los fármacos , Plasmodium chabaudi/efectos de los fármacos , Animales , Antimaláricos/aislamiento & purificación , Cloroquina/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Malaria/parasitología , Masculino , Ratones Endogámicos CBA , Parasitemia/tratamiento farmacológico , Parasitemia/parasitología , Pruebas de Sensibilidad Parasitaria , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Plantas Medicinales , Plasmodium berghei/patogenicidad , Plasmodium chabaudi/patogenicidad , Factores de Tiempo
18.
PLoS Biol ; 14(4): e1002436, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27088359

RESUMEN

Infected hosts differ in their responses to pathogens; some hosts are resilient and recover their original health, whereas others follow a divergent path and die. To quantitate these differences, we propose mapping the routes infected individuals take through "disease space." We find that when plotting physiological parameters against each other, many pairs have hysteretic relationships that identify the current location of the host and predict the future route of the infection. These maps can readily be constructed from experimental longitudinal data, and we provide two methods to generate the maps from the cross-sectional data that is commonly gathered in field trials. We hypothesize that resilient hosts tend to take small loops through disease space, whereas nonresilient individuals take large loops. We support this hypothesis with experimental data in mice infected with Plasmodium chabaudi, finding that dying mice trace a large arc in red blood cells (RBCs) by reticulocyte space as compared to surviving mice. We find that human malaria patients who are heterozygous for sickle cell hemoglobin occupy a small area of RBCs by reticulocyte space, suggesting this approach can be used to distinguish resilience in human populations. This technique should be broadly useful in describing the in-host dynamics of infections in both model hosts and patients at both population and individual levels.


Asunto(s)
Infecciones/fisiopatología , Animales , Eritrocitos , Humanos , Malaria/sangre , Malaria/fisiopatología , Ratones , Plasmodium chabaudi/patogenicidad
19.
Sci Rep ; 6: 23449, 2016 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-26996203

RESUMEN

Plasmodium multigene families play a central role in the pathogenesis of malaria. The Plasmodium interspersed repeat (pir) genes comprise the largest multigene family in many Plasmodium spp. However their function(s) remains unknown. Using the rodent model of malaria, Plasmodium chabaudi, we show that individual CIR proteins have differential localizations within infected red cell (iRBC), suggesting different functional roles in a blood-stage infection. Some CIRs appear to be located on the surface of iRBC and merozoites and are therefore well placed to interact with host molecules. In line with this hypothesis, we show for the first time that a subset of recombinant CIRs bind mouse RBCs suggesting a role for CIR in rosette formation and/or invasion. Together, our results unravel differences in subcellular localization and ability to bind mouse erythrocytes between the members of the cir family, which strongly suggest different functional roles in a blood-stage infection.


Asunto(s)
Eritrocitos/parasitología , Malaria/parasitología , Plasmodium chabaudi/genética , Plasmodium chabaudi/patogenicidad , Proteínas Protozoarias/fisiología , Animales , Genes Protozoarios , Secuencias Repetitivas Esparcidas , Malaria/genética , Ratones , Familia de Multigenes , Proteínas Protozoarias/biosíntesis
20.
Mediators Inflamm ; 2016: 3909614, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26903708

RESUMEN

The anemia and inflammation concurrent with blood stage malaria trigger stress haematopoiesis and erythropoiesis. The activity of osteoclasts seems required for the mobilization of hematopoietic stem and progenitor cells (HSPC) from the bone marrow to the periphery. Knowing that BALB/c mice with acute Plasmodium chabaudi adami malaria have profound alterations in bone remodelling cells, we evaluated the extent to which osteoclasts influence their hematopoietic response to infection. For this, mice were treated with osteoclast inhibiting hormone calcitonin prior to parasite inoculation, and infection as well as hematological parameters was studied. In agreement with osteoclast-dependent HSPC mobilization, administration of calcitonin led to milder splenomegaly, reduced numbers of HSPC in the spleen, and their retention in the bone marrow. Although C-terminal telopeptide (CTX) levels, indicative of bone resorption, were lower in calcitonin-treated infected mice, they remained comparable in naive and control infected mice. Calcitonin-treated infected mice conveniently responded to anemia but generated less numbers of splenic macrophages and suffered from exacerbated infection; interestingly, calcitonin also decreased the number of macrophages generated in vitro. Globally, our results indicate that although osteoclast-dependent HSC mobilization from bone marrow to spleen is triggered in murine blood stage malaria, this activity is not essential for stress erythropoiesis.


Asunto(s)
Movilización de Célula Madre Hematopoyética , Malaria/metabolismo , Osteoclastos/citología , Plasmodium chabaudi/patogenicidad , Células Madre/citología , Animales , Calcitonina/farmacología , Diferenciación Celular/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Osteoclastos/fisiología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA