Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Silvia Portugal is active.

Publication


Featured researches published by Silvia Portugal.


Nature Medicine | 2007

Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria

Ana Pamplona; Ana Ferreira; József Balla; Viktória Jeney; György Balla; Sabrina Epiphanio; Ângelo Chora; Isabel Pombo Gregoire; Margarida Cunha-Rodrigues; Silvia Portugal; Miguel P. Soares; Maria M. Mota

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.


Nature Medicine | 2011

Host-mediated regulation of superinfection in malaria

Silvia Portugal; Celine Carret; Mario Recker; Andrew E. Armitage; Lígia Antunes Gonçalves; Sabrina Epiphanio; David J. Sullivan; Cindy N. Roy; Chris Newbold; Hal Drakesmith; Maria M. Mota

In regions of high rates of malaria transmission, mosquitoes repeatedly transmit liver-tropic Plasmodium sporozoites to individuals who already have blood-stage parasitemia. This manifests itself in semi-immune children (who have been exposed since birth to Plasmodium infection and as such show low levels of peripheral parasitemia but can still be infected) older than 5 years of age by concurrent carriage of different parasite genotypes at low asymptomatic parasitemias. Superinfection presents an increased risk of hyperparasitemia and death in less immune individuals but counterintuitively is not frequently observed in the young. Here we show in a mouse model that ongoing blood-stage infections, above a minimum threshold, impair the growth of subsequently inoculated sporozoites such that they become growth arrested in liver hepatocytes and fail to develop into blood-stage parasites. Inhibition of the liver-stage infection is mediated by the host iron regulatory hormone hepcidin, whose synthesis we found to be stimulated by blood-stage parasites in a density-dependent manner. We mathematically modeled this phenomenon and show how density-dependent protection against liver-stage malaria can shape the epidemiological patterns of age-related risk and the complexity of malaria infections seen in young children. The interaction between these two Plasmodium stages and host iron metabolism has relevance for the global efforts to reduce malaria transmission and for evaluation of iron supplementation programs in malaria-endemic regions.


Annual Review of Immunology | 2014

Malaria Immunity in Man and Mosquito: Insights into Unsolved Mysteries of a Deadly Infectious Disease*

Peter D. Crompton; Jacqueline Moebius; Silvia Portugal; Michael Waisberg; Geoffrey T. Hart; Lindsey S. Garver; Louis H. Miller; Carolina Barillas-Mury; Susan K. Pierce

Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the worlds most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasites complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.


Cell Host & Microbe | 2008

Host Scavenger Receptor SR-BI Plays a Dual Role in the Establishment of Malaria Parasite Liver Infection

Michael Hannus; Miguel Prudêncio; Cécilie Martin; Lígia Antunes Gonçalves; Silvia Portugal; Sabrina Epiphanio; Akin Akinc; Philipp Hadwiger; Kerstin Jahn-Hofmann; Ingo Röhl; Geert-Jan van Gemert; Jean-François Franetich; Adrian J. F. Luty; Robert W. Sauerwein; Dominique Mazier; Victor Koteliansky; Hans-Peter Vornlocher; Christophe J. Echeverri; Maria M. Mota

An obligatory step of malaria parasite infection is Plasmodium sporozoite invasion of host hepatocytes, and host lipoprotein clearance pathways have been linked to Plasmodium liver infection. By using RNA interference to screen lipoprotein-related host factors, we show here that the class B, type I scavenger receptor (SR-BI) is the strongest regulator of Plasmodium infection among these factors. Inhibition of SR-BI function reduced P. berghei infection in Huh7 cells, and overexpression of SR-BI led to increased infection. In vivo silencing of liver SR-BI expression in mice and inhibition of SR-BI activity in human primary hepatocytes reduced infection by P. berghei and by P. falciparum, respectively. Heterozygous SR-BI(+/-) mice displayed reduced P. berghei infection rates correlating with liver SR-BI expression levels. Additional analyses revealed that SR-BI plays a dual role in Plasmodium infection, affecting both sporozoite invasion and intracellular parasite development, and may therefore constitute a good target for malaria prophylaxis.


Cell | 2014

Gut Microbiota Elicits a Protective Immune Response against Malaria Transmission

Bahtiyar Yilmaz; Silvia Portugal; Tuan M. Tran; Raffaella Gozzelino; Susana Ramos; Joana Gomes; Ana Regalado; Peter J. Cowan; Anthony J. F. D’Apice; Anita S. Chong; Ogobara K. Doumbo; Boubacar Traore; Peter D. Crompton; Henrique Silveira; Miguel P. Soares

Summary Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:β-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galβ1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans. PaperFlick


Cell Host & Microbe | 2008

Heme Oxygenase-1 Is an Anti-Inflammatory Host Factor that Promotes Murine Plasmodium Liver Infection

Sabrina Epiphanio; Sebastian A. Mikolajczak; Lígia Antunes Gonçalves; Ana Pamplona; Silvia Portugal; Sónia S. Albuquerque; Michael Goldberg; Sofia Rebelo; Daniel G. Anderson; Akin Akinc; Hans-Peter Vornlocher; Stefan H. I. Kappe; Miguel P. Soares; Maria M. Mota

The clinically silent Plasmodium liver stage is an obligatory step in the establishment of malaria infection and disease. We report here that expression of heme oxygenase-1 (HO-1, encoded by Hmox1) is upregulated in the liver following infection by Plasmodium berghei and Plasmodium yoelii sporozoites. HO-1 overexpression in the liver leads to a proportional increase in parasite liver load, and treatment of mice with carbon monoxide and with biliverdin, each an enzymatic product of HO-1, also increases parasite liver load. Conversely, mice lacking Hmox1 completely resolve the infection. In the absence of HO-1, the levels of inflammatory cytokines involved in the control of liver infection are increased. These findings suggest that, while stimulating inflammation, the liver stage of Plasmodium also induces HO-1 expression, which modulates the host inflammatory response, protecting the infected hepatocytes and promoting the liver stage of infection.


Journal of Immunology | 2013

Young Lives Lost as B Cells Falter: What We Are Learning About Antibody Responses in Malaria

Silvia Portugal; Susan K. Pierce; Peter D. Crompton

Plasmodium falciparum malaria remains a major public health threat for which there is no licensed vaccine. Abs play a key role in malaria immunity, but Ab-mediated protection is only acquired after years of repeated infections, leaving children in endemic areas vulnerable to severe malaria and death. Many P. falciparum Ags are extraordinarily diverse and clonally variant, which likely contribute to the inefficient acquisition of protective Abs. However, mounting evidence suggests that there is more to the story and that infection-induced dysregulation of B cell function also plays a role. We herein review progress toward understanding the B cell biology of P. falciparum infection, focusing on what has been learned from population-based studies in malaria-endemic areas. We suggest ways in which advances in immunology and genomics-based technology can further improve our understanding of the B cell response in malaria and perhaps illuminate new pathways to the development of effective vaccines.


PLOS Pathogens | 2014

Exposure-Dependent Control of Malaria-Induced Inflammation in Children

Silvia Portugal; Jacqueline Moebius; Jeff Skinner; Safiatou Doumbo; Didier Doumtabe; Younoussou Kone; Seydou Dia; Kishore Kanakabandi; Daniel E. Sturdevant; Kimmo Virtaneva; Stephen F. Porcella; Shanping Li; Ogobara K. Doumbo; Kassoum Kayentao; Aissata Ongoiba; Boubacar Traore; Peter D. Crompton

In malaria-naïve individuals, Plasmodium falciparum infection results in high levels of parasite-infected red blood cells (iRBCs) that trigger systemic inflammation and fever. Conversely, individuals in endemic areas who are repeatedly infected are often asymptomatic and have low levels of iRBCs, even young children. We hypothesized that febrile malaria alters the immune system such that P. falciparum re-exposure results in reduced production of pro-inflammatory cytokines/chemokines and enhanced anti-parasite effector responses compared to responses induced before malaria. To test this hypothesis we used a systems biology approach to analyze PBMCs sampled from healthy children before the six-month malaria season and the same children seven days after treatment of their first febrile malaria episode of the ensuing season. PBMCs were stimulated with iRBC in vitro and various immune parameters were measured. Before the malaria season, childrens immune cells responded to iRBCs by producing pro-inflammatory mediators such as IL-1β, IL-6 and IL-8. Following malaria there was a marked shift in the response to iRBCs with the same childrens immune cells producing lower levels of pro-inflammatory cytokines and higher levels of anti-inflammatory cytokines (IL-10, TGF-β). In addition, molecules involved in phagocytosis and activation of adaptive immunity were upregulated after malaria as compared to before. This shift was accompanied by an increase in P. falciparum-specific CD4+Foxp3− T cells that co-produce IL-10, IFN-γ and TNF; however, after the subsequent six-month dry season, a period of markedly reduced malaria transmission, P. falciparum–inducible IL-10 production remained partially upregulated only in children with persistent asymptomatic infections. These findings suggest that in the face of P. falciparum re-exposure, children acquire exposure-dependent P. falciparum–specific immunoregulatory responses that dampen pathogenic inflammation while enhancing anti-parasite effector mechanisms. These data provide mechanistic insight into the observation that P. falciparum–infected children in endemic areas are often afebrile and tend to control parasite replication.


eLife | 2015

Malaria-associated atypical memory B cells exhibit markedly reduced B cell receptor signaling and effector function

Silvia Portugal; Christopher Tipton; Haewon Sohn; Younoussou Kone; Jing Wang; Shanping Li; Jeff Skinner; Kimmo Virtaneva; Daniel E. Sturdevant; Stephen F. Porcella; Ogobara K. Doumbo; Safiatou Doumbo; Kassoum Kayentao; Aissata Ongoiba; Boubacar Traore; Ignacio Sanz; Susan K. Pierce; Peter D. Crompton

Protective antibodies in Plasmodium falciparum malaria are only acquired after years of repeated infections. Chronic malaria exposure is associated with a large increase in atypical memory B cells (MBCs) that resemble B cells expanded in a variety of persistent viral infections. Understanding the function of atypical MBCs and their relationship to classical MBCs will be critical to developing effective vaccines for malaria and other chronic infections. We show that VH gene repertoires and somatic hypermutation rates of atypical and classical MBCs are indistinguishable indicating a common developmental history. Atypical MBCs express an array of inhibitory receptors and B cell receptor (BCR) signaling is stunted in atypical MBCs resulting in impaired B cell responses including proliferation, cytokine production and antibody secretion. Thus, in response to chronic malaria exposure, atypical MBCs appear to differentiate from classical MBCs becoming refractory to BCR-mediated activation and potentially interfering with the acquisition of malaria immunity. DOI: http://dx.doi.org/10.7554/eLife.07218.001


Cell Reports | 2015

Circulating Th1-Cell-type Tfh Cells that Exhibit Impaired B Cell Help Are Preferentially Activated during Acute Malaria in Children.

Nyamekye Obeng-Adjei; Silvia Portugal; Tuan M. Tran; Takele Yazew; Jeff Skinner; Shanping Li; Aarti Jain; Philip L. Felgner; Ogobara K. Doumbo; Kassoum Kayentao; Aissata Ongoiba; Boubacar Traore; Peter D. Crompton

Malaria-specific antibody responses are short lived in children, leaving them susceptible to repeated bouts of febrile malaria. The cellular and molecular mechanisms underlying this apparent immune deficiency are poorly understood. Recently, T follicular helper (Tfh) cells have been shown to play a critical role in generating long-lived antibody responses. We show that Malian children have resting PD-1(+)CXCR5(+)CD4(+) Tfh cells in circulation that resemble germinal center Tfh cells phenotypically and functionally. Within this population, PD-1(+)CXCR5(+)CXCR3(-) Tfh cells are superior to Th1-polarized PD-1(+)CXCR5(+)CXCR3(+) Tfh cells in helping B cells. Longitudinally, we observed that malaria drives Th1 cytokine responses, and accordingly, the less-functional Th1-polarized Tfh subset was preferentially activated and its activation did not correlate with antibody responses. These data provide insights into the Tfh cell biology underlying suboptimal antibody responses to malaria in children and suggest that vaccine strategies that promote CXCR3(-) Tfh cell responses may improve malaria vaccine efficacy.

Collaboration


Dive into the Silvia Portugal's collaboration.

Top Co-Authors

Avatar

Peter D. Crompton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Maria M. Mota

Instituto de Medicina Molecular

View shared research outputs
Top Co-Authors

Avatar

Boubacar Traore

University of the Sciences

View shared research outputs
Top Co-Authors

Avatar

Jeff Skinner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tuan M. Tran

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Aissata Ongoiba

University of the Sciences

View shared research outputs
Top Co-Authors

Avatar

Shanping Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kassoum Kayentao

University of the Sciences

View shared research outputs
Top Co-Authors

Avatar

Susan K. Pierce

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge