Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 53(4): 733-744.e8, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32946741

RESUMEN

Discovering potent human monoclonal antibodies (mAbs) targeting the Plasmodium falciparum circumsporozoite protein (PfCSP) on sporozoites (SPZ) and elucidating their mechanisms of neutralization will facilitate translation for passive prophylaxis and aid next-generation vaccine development. Here, we isolated a neutralizing human mAb, L9 that preferentially bound NVDP minor repeats of PfCSP with high affinity while cross-reacting with NANP major repeats. L9 was more potent than six published neutralizing human PfCSP mAbs at mediating protection against mosquito bite challenge in mice. Isothermal titration calorimetry and multiphoton microscopy showed that L9 and the other most protective mAbs bound PfCSP with two binding events and mediated protection by killing SPZ in the liver and by preventing their egress from sinusoids and traversal of hepatocytes. This study defines the subdominant PfCSP minor repeats as neutralizing epitopes, identifies an in vitro biophysical correlate of SPZ neutralization, and demonstrates that the liver is an important site for antibodies to prevent malaria.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antiprotozoarios/inmunología , Antimaláricos/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Adolescente , Adulto , Animales , Línea Celular , Línea Celular Tumoral , Epítopos/inmunología , Femenino , Células HEK293 , Hepatocitos/inmunología , Hepatocitos/parasitología , Humanos , Hígado/inmunología , Hígado/parasitología , Malaria/inmunología , Malaria/parasitología , Vacunas contra la Malaria/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
2.
Proc Natl Acad Sci U S A ; 120(5): e2213626120, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36689648

RESUMEN

Plasmodium falciparum malaria originated when Plasmodium praefalciparum, a gorilla malaria parasite transmitted by African sylvan anopheline mosquitoes, adapted to humans. Pfs47, a protein on the parasite surface mediates P. falciparum evasion of the mosquito immune system by interacting with a midgut receptor and is critical for Plasmodium adaptation to different anopheline species. Genetic analysis of 4,971 Pfs47 gene sequences from different continents revealed that Asia and Papua New Guinea harbor Pfs47 haplotypes more similar to its ortholog in P. praefalciparum at sites that determine vector compatibility, suggesting that ancestral P. falciparum readily adapted to Asian vectors. Consistent with this observation, Pfs47-receptor gene sequences from African sylvan malaria vectors, such as Anopheles moucheti and An. marshallii, were found to share greater similarity with those of Asian vectors than those of vectors of the African An. gambiae complex. Furthermore, experimental infections provide direct evidence that transformed P. falciparum parasites carrying Pfs47 orthologs of P. praefalciparum or P. reichenowi were more effective at evading the immune system of the Asian malaria vector An. dirus than An. gambiae. We propose that high compatibility of ancestral P. falciparum Pfs47 with the receptors of Asian vectors facilitated the early dispersal of human malaria to the Asian continent, without having to first adapt to sub-Saharan vectors of the An. gambiae complex.


Asunto(s)
Anopheles , Malaria Falciparum , Malaria , Plasmodium , Animales , Humanos , Plasmodium falciparum/genética , Anopheles/genética , Mosquitos Vectores/parasitología , Malaria Falciparum/parasitología , Gorilla gorilla
3.
Proc Natl Acad Sci U S A ; 119(35): e2209729119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994647

RESUMEN

Glutaminyl cyclase (QC) modifies N-terminal glutamine or glutamic acid residues of target proteins into cyclic pyroglutamic acid (pGlu). Here, we report the biochemical and functional analysis of Plasmodium QC. We show that sporozoites of QC-null mutants of rodent and human malaria parasites are recognized by the mosquito immune system and melanized when they reach the hemocoel. Detailed analyses of rodent malaria QC-null mutants showed that sporozoite numbers in salivary glands are reduced in mosquitoes infected with QC-null or QC catalytically dead mutants. This phenotype can be rescued by genetic complementation or by disrupting mosquito melanization or phagocytosis by hemocytes. Mutation of a single QC-target glutamine of the major sporozoite surface protein (circumsporozoite protein; CSP) of the rodent parasite Plasmodium berghei also results in melanization of sporozoites. These findings indicate that QC-mediated posttranslational modification of surface proteins underlies evasion of killing of sporozoites by the mosquito immune system.


Asunto(s)
Aminoaciltransferasas , Culicidae , Malaria , Procesamiento Proteico-Postraduccional , Esporozoítos , Aminoaciltransferasas/inmunología , Animales , Culicidae/inmunología , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Malaria/genética , Malaria/inmunología , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología
4.
Annu Rev Genet ; 50: 447-465, 2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-27732796

RESUMEN

Plasmodium falciparum malaria remains a devastating public health problem. Recent discoveries have shed light on the origin and evolution of Plasmodium parasites and their interactions with their vertebrate and mosquito hosts. P. falciparum malaria originated in Africa from a single horizontal transfer between an infected gorilla and a human, and became global as the result of human migration. Today, P. falciparum malaria is transmitted worldwide by more than 70 different anopheline mosquito species. Recent studies indicate that the mosquito immune system can be a barrier to malaria transmission and that the P. falciparum Pfs47 gene allows the parasite to evade mosquito immune detection. Here, we review the origin and globalization of P. falciparum and integrate this history with analysis of the biology, evolution, and dispersal of the main mosquito vectors. This new perspective broadens our understanding of P. falciparum population structure and the dispersal of important parasite genetic traits.


Asunto(s)
Malaria Falciparum/transmisión , Mosquitos Vectores , Plasmodium falciparum/patogenicidad , Adaptación Biológica/genética , Animales , Anopheles/parasitología , Variación Genética , Humanos , Evasión Inmune , Mosquitos Vectores/inmunología , Mosquitos Vectores/patogenicidad , Plasmodium falciparum/genética
5.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34711682

RESUMEN

Immune priming in Anopheles gambiae is mediated by the systemic release of a hemocyte differentiation factor (HDF), a complex of lipoxin A4 bound to Evokin, a lipid carrier. HDF increases the proportion of circulating granulocytes and enhances mosquito cellular immunity. Here, we show that Evokin is present in hemocytes and fat-body cells, and messenger RNA (mRNA) expression increases significantly after immune priming. The double peroxidase (DBLOX) enzyme, present in insects but not in vertebrates, is essential for HDF synthesis. DBLOX is highly expressed in oenocytes in the fat-body tissue, and these cells increase in number in primed mosquitoes. We provide direct evidence that the histone acetyltransferase AgTip60 (AGAP001539) is also essential for a sustained increase in oenocyte numbers, HDF synthesis, and immune priming. We propose that oenocytes may function as a population of cells that are reprogrammed, and orchestrate and maintain a broad, systemic, and long-lasting state of enhanced immune surveillance in primed mosquitoes.


Asunto(s)
Culicidae/inmunología , Histona Acetiltransferasas/metabolismo , Memoria Inmunológica/inmunología , Animales , Anopheles/inmunología , Anopheles/metabolismo , Culicidae/metabolismo , Femenino , Granulocitos/metabolismo , Hemocitos/inmunología , Inmunidad Innata/inmunología , Proteínas de Insectos/genética , Insectos/metabolismo , Lipoxinas/metabolismo , Malaria/inmunología , Masculino , Peroxidasa/metabolismo , Plasmodium/metabolismo , Plasmodium berghei/metabolismo
6.
PLoS Pathog ; 17(12): e1010133, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34871332

RESUMEN

Combinations of monoclonal antibodies (mAbs) against different epitopes on the same antigen synergistically neutralize many viruses. However, there are limited studies assessing whether combining human mAbs against distinct regions of the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) enhances in vivo protection against malaria compared to each mAb alone or whether passive transfer of PfCSP mAbs would improve protection following vaccination against PfCSP. Here, we isolated a panel of human mAbs against the subdominant C-terminal domain of PfCSP (C-CSP) from a volunteer immunized with radiation-attenuated Pf sporozoites. These C-CSP-specific mAbs had limited binding to sporozoites in vitro that was increased by combination with neutralizing human "repeat" mAbs against the NPDP/NVDP/NANP tetrapeptides in the central repeat region of PfCSP. Nevertheless, passive transfer of repeat- and C-CSP-specific mAb combinations did not provide enhanced protection against in vivo sporozoite challenge compared to repeat mAbs alone. Furthermore, combining potent repeat-specific mAbs (CIS43, L9, and 317) that respectively target the three tetrapeptides (NPDP/NVDP/NANP) did not provide additional protection against in vivo sporozoite challenge. However, administration of either CIS43, L9, or 317 (but not C-CSP-specific mAbs) to mice that had been immunized with R21, a PfCSP-based virus-like particle vaccine that induces polyclonal antibodies against the repeat region and C-CSP, provided enhanced protection against sporozoite challenge when compared to vaccine or mAbs alone. Collectively, this study shows that while combining mAbs against the repeat and C-terminal regions of PfCSP provide no additional protection in vivo, repeat mAbs do provide increased protection when combined with vaccine-induced polyclonal antibodies. These data should inform the implementation of PfCSP human mAbs alone or following vaccination to prevent malaria infection.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunización Pasiva/métodos , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Humanos , Malaria Falciparum/prevención & control , Ratones , Esporozoítos/inmunología
7.
Proc Natl Acad Sci U S A ; 117(30): 17720-17726, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32647056

RESUMEN

A series of epidemiological explorations has suggested a negative association between national bacillus Calmette-Guérin (BCG) vaccination policy and the prevalence and mortality of coronavirus disease 2019 (COVID-19). However, these comparisons are difficult to validate due to broad differences between countries such as socioeconomic status, demographic structure, rural vs. urban settings, time of arrival of the pandemic, number of diagnostic tests and criteria for testing, and national control strategies to limit the spread of COVID-19. We review evidence for a potential biological basis of BCG cross-protection from severe COVID-19, and refine the epidemiological analysis to mitigate effects of potentially confounding factors (e.g., stage of the COVID-19 epidemic, development, rurality, population density, and age structure). A strong correlation between the BCG index, an estimation of the degree of universal BCG vaccination deployment in a country, and COVID-19 mortality in different socially similar European countries was observed (r2 = 0.88; P = 8 × 10-7), indicating that every 10% increase in the BCG index was associated with a 10.4% reduction in COVID-19 mortality. Results fail to confirm the null hypothesis of no association between BCG vaccination and COVID-19 mortality, and suggest that BCG could have a protective effect. Nevertheless, the analyses are restricted to coarse-scale signals and should be considered with caution. BCG vaccination clinical trials are required to corroborate the patterns detected here, and to establish causality between BCG vaccination and protection from severe COVID-19. Public health implications of a plausible BCG cross-protection from severe COVID-19 are discussed.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacuna BCG/administración & dosificación , Betacoronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/mortalidad , Neumonía Viral/prevención & control , Anciano , Betacoronavirus/efectos de los fármacos , COVID-19 , Infecciones por Coronavirus/virología , Humanos , Neumonía Viral/virología , Pronóstico , SARS-CoV-2 , Tasa de Supervivencia , Vacunación
8.
Proc Natl Acad Sci U S A ; 117(5): 2597-2605, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31969456

RESUMEN

The surface protein Pfs47 allows Plasmodium falciparum parasites to survive and be transmitted by making them "undetectable" to the mosquito immune system. P. falciparum parasites express Pfs47 haplotypes compatible with their sympatric vectors, while those with incompatible haplotypes are eliminated by the mosquito. We proposed that Pfs47 serves as a "key" that mediates immune evasion by interacting with a mosquito receptor "the lock," which differs in evolutionarily divergent anopheline mosquitoes. Recombinant Pfs47 (rPfs47) was used to identify the mosquito Pfs47 receptor protein (P47Rec) using far-Western analysis. rPfs47 bound to a single 31-kDa band and the identity of this protein was determined by mass spectrometry. The mosquito P47Rec has two natterin-like domains and binds to Pfs47 with high affinity (17 to 32 nM). P47Rec is a highly conserved protein with submicrovillar localization in midgut cells. It has structural homology to a cytoskeleton-interacting protein and accumulates at the site of ookinete invasion. Silencing P47Rec expression reduced P. falciparum infection, indicating that the interaction of Pfs47 with the receptor is critical for parasite survival. The binding specificity of P47Rec from distant anophelines (Anopheles gambiae, Anopheles dirus, and Anopheles albimanus) with Pfs47-Africa (GB4) and Pfs47-South America (7G8) haplotypes was evaluated, and it is in agreement with the previously documented compatibility between P. falciparum parasites expressing different Pfs47 haplotypes and these three anopheline species. Our findings give further support to the role of Pfs47 in the adaptation of P. falciparum to different vectors.


Asunto(s)
Anopheles/inmunología , Anopheles/parasitología , Proteínas de Insectos/inmunología , Glicoproteínas de Membrana/inmunología , Mosquitos Vectores/inmunología , Mosquitos Vectores/parasitología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anopheles/genética , Interacciones Huésped-Parásitos , Evasión Inmune , Proteínas de Insectos/genética , Cinética , Glicoproteínas de Membrana/genética , Mosquitos Vectores/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética
9.
Malar J ; 20(1): 11, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407511

RESUMEN

BACKGROUND: The invasion of the mosquito salivary glands by Plasmodium sporozoites is a critical step that defines the success of malaria transmission and a detailed understanding of the molecules responsible for salivary gland invasion could be leveraged towards control of vector-borne pathogens. Antibodies directed against the mosquito salivary gland protein SGS1 have been shown to reduce Plasmodium gallinaceum sporozoite invasion of Aedes aegypti salivary glands, but the specific role of this protein in sporozoite invasion and in other stages of the Plasmodium life cycle remains unknown. METHODS: RNA interference and CRISPR/Cas9 were used to evaluate the role of A. aegypti SGS1 in the P. gallinaceum life cycle. RESULTS: Knockdown and knockout of SGS1 disrupted sporozoite invasion of the salivary gland. Interestingly, mosquitoes lacking SGS1 also displayed fewer oocysts. Proteomic analyses confirmed the abolishment of SGS1 in the salivary gland of SGS1 knockout mosquitoes and revealed that the C-terminus of the protein is absent in the salivary gland of control mosquitoes. In silico analyses indicated that SGS1 contains two potential internal cleavage sites and thus might generate three proteins. CONCLUSION: SGS1 facilitates, but is not essential for, invasion of A. aegypti salivary glands by P. gallinaceum and has a dual role as a facilitator of parasite development in the mosquito midgut. SGS1 could, therefore, be part of a strategy to decrease malaria transmission by the mosquito vector, for example in a transgenic mosquito that blocks its interaction with the parasite.


Asunto(s)
Aedes/genética , Proteínas de Insectos/genética , Plasmodium gallinaceum/fisiología , Proteínas y Péptidos Salivales/genética , Aedes/parasitología , Secuencia de Aminoácidos , Animales , Femenino , Tracto Gastrointestinal/parasitología , Proteínas de Insectos/química , Proteínas de Insectos/metabolismo , Mosquitos Vectores/genética , Mosquitos Vectores/parasitología , Glándulas Salivales/parasitología , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/metabolismo , Alineación de Secuencia , Esporozoítos/fisiología
10.
Proc Natl Acad Sci U S A ; 115(4): 774-779, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311293

RESUMEN

Efforts to knock out Plasmodium falciparum calcium-dependent protein kinase 1 (PfCDPK1) from asexual erythrocytic stage have not been successful, indicating an indispensable role of the enzyme in asexual growth. We recently reported generation of a transgenic parasite with mutant CDPK1 [Bansal A, et al. (2016) MBio 7:e02011-16]. The mutant CDPK1 (T145M) had reduced activity of transphosphorylation. We reasoned that CDPK1 could be disrupted in the mutant parasites. Consistent with this assumption, CDPK1 was successfully disrupted in the mutant parasites using CRISPR/Cas9. We and others could not disrupt PfCDPK1 in the WT parasites. The CDPK1 KO parasites show a slow growth rate compared with the WT and the CDPK1 T145M parasites. Additionally, the CDPK1 KO parasites show a defect in both male and female gametogenesis and could not establish an infection in mosquitoes. Complementation of the KO parasite with full-length PfCDPK1 partially rescued the asexual growth defect and mosquito infection. Comparative global transcriptomics of WT and the CDPK1 KO schizonts using RNA-seq show significantly high transcript expression of gametocyte-specific genes in the CDPK1 KO parasites. This study conclusively demonstrates that CDPK1 is a good target for developing transmission-blocking drugs.


Asunto(s)
Culicidae/parasitología , Gametogénesis , Proteínas Quinasas/fisiología , Proteínas Protozoarias/fisiología , Animales , Sistemas CRISPR-Cas , Edición Génica , Regulación de la Expresión Génica , Plasmodium falciparum
11.
Proc Natl Acad Sci U S A ; 114(47): 12566-12571, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29114059

RESUMEN

A naturally occurring Wolbachia strain (wAnga-Mali) was identified in mosquitoes of the Anopheles gambiae complex collected in the Malian villages of Dangassa and Kenieroba. Phylogenetic analysis of the nucleotide sequence of two 16S rRNA regions showed that wAnga-Mali clusters with Wolbachia strains from supergroup A and has the highest homology to a Wolbachia strain isolated from cat fleas (Ctenocephalides). wAnga-Mali is different from two Wolbachia strains previously reported in A. gambiae from Burkina Faso (wAnga_VK5_STP and wAnga_VK5_3.1a). Quantitative analysis of Wolbachia and Plasmodium sporozoite infection in field-collected mosquitoes indicates that the prevalence and intensity of Plasmodium falciparum sporozoite infection is significantly lower in Wolbachia-infected females. The presence of Wolbachia in females from a laboratory Anopheles coluzzii (A. gambiae, M form) colony experimentally infected with P. falciparum (NF54 strain) gametocyte cultures slightly enhanced oocyst infection. However, Wolbachia infection significantly reduced the prevalence and intensity of sporozoite infection, as observed in the field. This indicates that wAnga-Mali infection does not limit early stages of Plasmodium infection in the mosquito, but it has a strong deleterious effect on sporozoites and reduces malaria transmission.


Asunto(s)
Anopheles/microbiología , Interacciones Huésped-Parásitos , Insectos Vectores/microbiología , Malaria Falciparum/transmisión , Plasmodium falciparum/microbiología , Wolbachia/genética , Animales , Anopheles/parasitología , Femenino , Interacciones Huésped-Patógeno , Insectos Vectores/parasitología , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Malí/epidemiología , Oocistos/patogenicidad , Oocistos/fisiología , Filogenia , ARN Ribosómico 16S/genética , Índice de Severidad de la Enfermedad , Esporozoítos/patogenicidad , Esporozoítos/fisiología , Wolbachia/clasificación , Wolbachia/aislamiento & purificación
14.
Proc Natl Acad Sci U S A ; 112(5): 1273-80, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25552553

RESUMEN

The malaria parasite, Plasmodium, must survive and develop in the mosquito vector to be successfully transmitted to a new host. The Plasmodium falciparum Pfs47 gene is critical for malaria transmission. Parasites that express Pfs47 (NF54 WT) evade mosquito immunity and survive, whereas Pfs47 knockouts (KO) are efficiently eliminated by the complement-like system. Two alternative approaches were used to investigate the mechanism of action of Pfs47 on immune evasion. First, we examined whether Pfs47 affected signal transduction pathways mediating mosquito immune responses, and show that the Jun-N-terminal kinase (JNK) pathway is a key mediator of Anopheles gambiae antiplasmodial responses to P. falciparum infection and that Pfs47 disrupts JNK signaling. Second, we used microarrays to compare the global transcriptional responses of A. gambiae midguts to infection with WT and KO parasites. The presence of Pfs47 results in broad and profound changes in gene expression in response to infection that are already evident 12 h postfeeding, but become most prominent at 26 h postfeeding, the time when ookinetes invade the mosquito midgut. Silencing of 15 differentially expressed candidate genes identified caspase-S2 as a key effector of Plasmodium elimination in parasites lacking Pfs47. We provide experimental evidence that JNK pathway regulates activation of caspases in Plasmodium-invaded midgut cells, and that caspase activation is required to trigger midgut epithelial nitration. Pfs47 alters the cell death pathway of invaded midgut cells by disrupting JNK signaling and prevents the activation of several caspases, resulting in an ineffective nitration response that makes the parasite undetectable by the mosquito complement-like system.


Asunto(s)
Anopheles/inmunología , Apoptosis/fisiología , MAP Quinasa Quinasa 4/metabolismo , Plasmodium falciparum/fisiología , Animales , Anopheles/parasitología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/fisiología
15.
Proc Natl Acad Sci U S A ; 112(49): 15178-83, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598665

RESUMEN

Plasmodium falciparum malaria originated in Africa and became global as humans migrated to other continents. During this journey, parasites encountered new mosquito species, some of them evolutionarily distant from African vectors. We have previously shown that the Pfs47 protein allows the parasite to evade the mosquito immune system of Anopheles gambiae mosquitoes. Here, we investigated the role of Pfs47-mediated immune evasion in the adaptation of P. falciparum to evolutionarily distant mosquito species. We found that P. falciparum isolates from Africa, Asia, or the Americas have low compatibility to malaria vectors from a different continent, an effect that is mediated by the mosquito immune system. We identified 42 different haplotypes of Pfs47 that have a strong geographic population structure and much lower haplotype diversity outside Africa. Replacement of the Pfs47 haplotypes in a P. falciparum isolate is sufficient to make it compatible to a different mosquito species. Those parasites that express a Pfs47 haplotype compatible with a given vector evade antiplasmodial immunity and survive. We propose that Pfs47-mediated immune evasion has been critical for the globalization of P. falciparum malaria as parasites adapted to new vector species. Our findings predict that this ongoing selective force by the mosquito immune system could influence the dispersal of Plasmodium genetic traits and point to Pfs47 as a potential target to block malaria transmission. A new model, the "lock-and-key theory" of P. falciparum globalization, is proposed, and its implications are discussed.


Asunto(s)
Anopheles/inmunología , Evasión Inmune , Malaria Falciparum/transmisión , Plasmodium falciparum/fisiología , Animales , Anopheles/parasitología , Insectos Vectores , Datos de Secuencia Molecular
16.
PLoS Pathog ; 10(9): e1004338, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25211214

RESUMEN

BACKGROUND: Invasion of mosquito salivary glands (SGs) by Plasmodium falciparum sporozoites is an essential step in the malaria life cycle. How infection modulates gene expression, and affects hematophagy remains unclear. PRINCIPAL FINDINGS: Using Affimetrix chip microarray, we found that at least 43 genes are differentially expressed in the glands of Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Among the upregulated genes, one codes for Agaphelin, a 58-amino acid protein containing a single Kazal domain with a Leu in the P1 position. Agaphelin displays high homology to orthologs present in Aedes sp and Culex sp salivary glands, indicating an evolutionarily expanded family. Kinetics and surface plasmon resonance experiments determined that chemically synthesized Agaphelin behaves as a slow and tight inhibitor of neutrophil elastase (K(D) ∼ 10 nM), but does not affect other enzymes, nor promotes vasodilation, or exhibit antimicrobial activity. TAXIscan chamber assay revealed that Agaphelin inhibits neutrophil chemotaxis toward fMLP, affecting several parameter associated with cell migration. In addition, Agaphelin reduces paw edema formation and accumulation of tissue myeloperoxidase triggered by injection of carrageenan in mice. Agaphelin also blocks elastase/cathepsin-mediated platelet aggregation, abrogates elastase-mediated cleavage of tissue factor pathway inhibitor, and attenuates neutrophil-induced coagulation. Notably, Agaphelin inhibits neutrophil extracellular traps (NETs) formation and prevents FeCl3-induced arterial thrombosis, without impairing hemostasis. CONCLUSIONS: Blockade of neutrophil elastase emerges as a novel antihemostatic mechanism in hematophagy; it also supports the notion that neutrophils and the innate immune response are targets for antithrombotic therapy. In addition, Agaphelin is the first antihemostatic whose expression is induced by Plasmodium sp infection. These results suggest that an important interplay takes place in parasite-vector-host interactions.


Asunto(s)
Anopheles/parasitología , Hemostasis/fisiología , Interacciones Huésped-Parásitos , Proteínas de Insectos/metabolismo , Neutrófilos/inmunología , Plasmodium falciparum/patogenicidad , Proteínas y Péptidos Salivales/metabolismo , Trombosis/prevención & control , Secuencia de Aminoácidos , Animales , Anopheles/metabolismo , Dicroismo Circular , Edema/etiología , Edema/metabolismo , Edema/prevención & control , Femenino , Proteínas de Insectos/química , Proteínas de Insectos/genética , Insectos Vectores , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Glándulas Salivales/metabolismo , Glándulas Salivales/parasitología , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/genética , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
17.
Malar J ; 15(1): 394, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27480269

RESUMEN

BACKGROUND: Malaria is transmitted when an infected mosquito delivers Plasmodium sporozoites into a vertebrate host. There are many species of Plasmodium and, in general, the infection is host-specific. For example, Plasmodium gallinaceum is an avian parasite, while Plasmodium berghei infects mice. These two parasites have been extensively used as experimental models of malaria transmission. Plasmodium falciparum and Plasmodium vivax are the most important agents of human malaria, a life-threatening disease of global importance. To complete their life cycle, Plasmodium parasites must traverse the mosquito midgut and form an oocyst that will divide continuously. Mature oocysts release thousands of sporozoites into the mosquito haemolymph that must reach the salivary gland to infect a new vertebrate host. The current understanding of the biology of oocyst formation and sporozoite release is mostly based on experimental infections with P. berghei, and the conclusions are generalized to other Plasmodium species that infect humans without further morphological analyses. RESULTS: Here, it is described the microanatomy of sporozoite escape from oocysts of four Plasmodium species: the two laboratory models, P. gallinaceum and P. berghei, and the two main species that cause malaria in humans, P. vivax and P. falciparum. It was found that sporozoites have species-specific mechanisms of escape from the oocyst. The two model species of Plasmodium had a common mechanism, in which the oocyst wall breaks down before sporozoites emerge. In contrast, P. vivax and P. falciparum sporozoites show a dynamic escape mechanism from the oocyst via polarized propulsion. CONCLUSIONS: This study demonstrated that Plasmodium species do not share a common mechanism of sporozoite escape, as previously thought, but show complex and species-specific mechanisms. In addition, the knowledge of this phenomenon in human Plasmodium can facilitate transmission-blocking studies and not those ones only based on the murine and avian models.


Asunto(s)
Oocistos/parasitología , Oocistos/ultraestructura , Plasmodium/fisiología , Plasmodium/ultraestructura , Esporozoítos/fisiología , Esporozoítos/ultraestructura , Animales , Aves , Femenino , Humanos , Estadios del Ciclo de Vida , Ratones , Microscopía Electrónica de Rastreo
18.
Infect Immun ; 83(10): 3771-80, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26169272

RESUMEN

The extended rod-like Plasmodium falciparum circumsporozoite protein (CSP) is comprised of three primary domains: a charged N terminus that binds heparan sulfate proteoglycans, a central NANP repeat domain, and a C terminus containing a thrombospondin-like type I repeat (TSR) domain. Only the last two domains are incorporated in RTS,S, the leading malaria vaccine in phase 3 trials that, to date, protects about 50% of vaccinated children against clinical disease. A seroepidemiological study indicated that the N-terminal domain might improve the efficacy of a new CSP vaccine. Using a panel of CSP-specific monoclonal antibodies, well-characterized recombinant CSPs, label-free quantitative proteomics, and in vitro inhibition of sporozoite invasion, we show that native CSP is N-terminally processed in the mosquito host and undergoes a reversible conformational change to mask some epitopes in the N- and C-terminal domains until the sporozoite interacts with the liver hepatocyte. Our findings show the importance of understanding processing and the biophysical change in conformation, possibly due to a mechanical or molecular signal, and may aid in the development of a new CSP vaccine.


Asunto(s)
Malaria Falciparum/parasitología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Animales , Anopheles/parasitología , Anticuerpos Antiprotozoarios/inmunología , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Hepatocitos/inmunología , Hepatocitos/parasitología , Humanos , Malaria Falciparum/inmunología , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/genética , Esporozoítos/química , Esporozoítos/crecimiento & desarrollo
19.
Proc Natl Acad Sci U S A ; 109(28): E1957-62, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22623529

RESUMEN

Plasmodium falciparum lines differ in their ability to infect mosquitoes. The Anopheles gambiae L3-5 refractory (R) line melanizes most Plasmodium species, including the Brazilian P. falciparum 7G8 line, but it is highly susceptible to some African P. falciparum strains such as 3D7, NF54, and GB4. We investigated whether these lines differ in their ability to evade the mosquito immune system. Silencing key components of the mosquito complement-like system [thioester-containing protein 1 (TEP1), leucine-rich repeat protein 1, and Anopheles Plasmodium-responsive leucine-rich repeat protein 1] prevented melanization of 7G8 parasites, reverting the refractory phenotype. In contrast, it had no effect on the intensity of infection with NF54, suggesting that this line is able to evade TEP1-mediated lysis. When R females were coinfected with a line that is melanized (7G8) and a line that survives (3D7), the coinfection resulted in mixed infections with both live and encapsulated parasites on individual midguts. This finding shows that survival of individual parasites is parasite-specific and not systemic in nature, because parasites can evade TEP1-mediated lysis even when other parasites are melanized in the same midgut. When females from an extensive genetic cross between R and susceptible A. gambiae (G3) mosquitoes were infected with P. berghei, encapsulation was strongly correlated with the TEP1-R1 allele. However, P. falciparum 7G8 parasites were no longer encapsulated by females from this cross, indicating that the TEP1-R1 allele is not sufficient to melanize this line. Evasion of the A. gambiae immune system by P. falciparum may be the result of parasite adaptation to sympatric mosquito vectors and may be an important factor driving malaria transmission.


Asunto(s)
Anopheles/metabolismo , Anopheles/parasitología , Proteínas del Sistema Complemento/metabolismo , Proteínas de Insectos/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Alelos , Animales , Cruzamientos Genéticos , Femenino , Humanos , Sistema Inmunológico , Malaria/parasitología , Malaria/transmisión , ARN Bicatenario/genética , Especificidad de la Especie
20.
BMC Genomics ; 15: 636, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25073905

RESUMEN

BACKGROUND: Genome sequencing of Anopheles gambiae was completed more than ten years ago and has accelerated research on malaria transmission. However, annotation needs to be refined and verified experimentally, as most predicted transcripts have been identified by comparative analysis with genomes from other species. The mosquito midgut-the first organ to interact with Plasmodium parasites-mounts effective antiplasmodial responses that limit parasite survival and disease transmission. High-throughput Illumina sequencing of the midgut transcriptome was used to identify new genes and transcripts, contributing to the refinement of An. gambiae genome annotation. RESULTS: We sequenced ~223 million reads from An. gambiae midgut cDNA libraries generated from susceptible (G3) and refractory (L35) mosquito strains. Mosquitoes were infected with either Plasmodium berghei or Plasmodium falciparum, and midguts were collected after the first or second Plasmodium infection. In total, 22,889 unique midgut transcript models were generated from both An. gambiae strain sequences combined, and 76% are potentially novel. Of these novel transcripts, 49.5% aligned with annotated genes and appear to be isoforms or pre-mRNAs of reference transcripts, while 50.5% mapped to regions between annotated genes and represent novel intergenic transcripts (NITs). Predicted models were validated for midgut expression using qRT-PCR and microarray analysis, and novel isoforms were confirmed by sequencing predicted intron-exon boundaries. Coding potential analysis revealed that 43% of total midgut transcripts appear to be long non-coding RNA (lncRNA), and functional annotation of NITs showed that 68% had no homology to current databases from other species. Reads were also analyzed using de novo assembly and predicted transcripts compared with genome mapping-based models. Finally, variant analysis of G3 and L35 midgut transcripts detected 160,742 variants with respect to the An. gambiae PEST genome, and 74% were new variants. Intergenic transcripts had a higher frequency of variation compared with non-intergenic transcripts. CONCLUSION: This in-depth Illumina sequencing and assembly of the An. gambiae midgut transcriptome doubled the number of known transcripts and tripled the number of variants known in this mosquito species. It also revealed existence of a large number of lncRNA and opens new possibilities for investigating the biological function of many newly discovered transcripts.


Asunto(s)
Anopheles/genética , Mucosa Intestinal/metabolismo , Anotación de Secuencia Molecular/métodos , Transcriptoma , Animales , Anopheles/embriología , Anopheles/parasitología , Variación Genética , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Plasmodium berghei/fisiología , Plasmodium falciparum/fisiología , ARN Mensajero/genética , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA