Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
PLoS Pathog ; 10(6): e1004231, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24968248

RESUMO

Insulin and insulin-like growth factor signaling (IIS) regulates cell death, repair, autophagy, and renewal in response to stress, damage, and pathogen challenge. Therefore, IIS is fundamental to lifespan and disease resistance. Previously, we showed that insulin-like growth factor 1 (IGF1) within a physiologically relevant range (0.013-0.13 µM) in human blood reduced development of the human parasite Plasmodium falciparum in the Indian malaria mosquito Anopheles stephensi. Low IGF1 (0.013 µM) induced FOXO and p70S6K activation in the midgut and extended mosquito lifespan, whereas high IGF1 (0.13 µM) did not. In this study the physiological effects of low and high IGF1 were examined in detail to infer mechanisms for their dichotomous effects on mosquito resistance and lifespan. Following ingestion, low IGF1 induced phosphorylation of midgut c-Jun-N-terminal kinase (JNK), a critical regulator of epithelial homeostasis, but high IGF1 did not. Low and high IGF1 induced midgut mitochondrial reactive oxygen species (ROS) synthesis and nitric oxide (NO) synthase gene expression, responses which were necessary and sufficient to mediate IGF1 inhibition of P. falciparum development. However, increased ROS and apoptosis-associated caspase-3 activity returned to baseline levels following low IGF1 treatment, but were sustained with high IGF1 treatment and accompanied by aberrant expression of biomarkers for mitophagy, stem cell division and proliferation. Low IGF1-induced ROS are likely moderated by JNK-induced epithelial cytoprotection as well as p70S6K-mediated growth and inhibition of apoptosis over the lifetime of A. stephensi to facilitate midgut homeostasis and enhanced survivorship. Hence, mitochondrial integrity and homeostasis in the midgut, a key signaling center for IIS, can be targeted to coordinately optimize mosquito fitness and anti-pathogen resistance for improved control strategies for malaria and other vector-borne diseases.


Assuntos
Anopheles/efeitos dos fármacos , Interações Hospedeiro-Parasita/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Plasmodium falciparum/patogenicidade , Animais , Anopheles/crescimento & desenvolvimento , Anopheles/metabolismo , Anopheles/parasitologia , Controle de Doenças Transmissíveis , Feminino , Homeostase/efeitos dos fármacos , Hormese , Humanos , Proteínas de Insetos/metabolismo , Insetos Vetores/efeitos dos fármacos , Insetos Vetores/crescimento & desenvolvimento , Insetos Vetores/metabolismo , Insetos Vetores/parasitologia , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/genética , Mucosa Intestinal/metabolismo , Longevidade/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fosforilação/efeitos dos fármacos , Plasmodium falciparum/isolamento & purificação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
2.
PLoS Pathog ; 9(2): e1003180, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23468624

RESUMO

The overexpression of activated, myristoylated Akt in the midgut of female transgenic Anopheles stephensi results in resistance to infection with the human malaria parasite Plasmodium falciparum but also decreased lifespan. In the present study, the understanding of mitochondria-dependent midgut homeostasis has been expanded to explain this apparent paradox in an insect of major medical importance. Given that Akt signaling is essential for cell growth and survival, we hypothesized that sustained Akt activation in the mosquito midgut would alter the balance of critical pathways that control mitochondrial dynamics to enhance parasite killing at some cost to survivorship. Toxic reactive oxygen and nitrogen species (RNOS) rise to high levels in the midgut after blood feeding, due to a combination of high NO production and a decline in FOXO-dependent antioxidants. Despite an apparent increase in mitochondrial biogenesis in young females (3 d), energy deficiencies were apparent as decreased oxidative phosphorylation and increased [AMP]/[ATP] ratios. In addition, mitochondrial mass was lower and accompanied by the presence of stalled autophagosomes in the posterior midgut, a critical site for blood digestion and stem cell-mediated epithelial maintenance and repair, and by functional degradation of the epithelial barrier. By 18 d, the age at which An. stephensi would transmit P. falciparum to human hosts, mitochondrial dysfunction coupled to Akt-mediated repression of autophagy/mitophagy was more evident and midgut epithelial structure was markedly compromised. Inhibition of RNOS by co-feeding of the nitric-oxide synthase inhibitor L-NAME at infection abrogated Akt-dependent killing of P. falciparum that begins within 18 h of infection in 3-5 d old mosquitoes. Hence, Akt-induced changes in mitochondrial dynamics perturb midgut homeostasis to enhance parasite resistance and decrease mosquito infective lifespan. Further, quality control of mitochondrial function in the midgut is necessary for the maintenance of midgut health as reflected in energy homeostasis and tissue repair and renewal.


Assuntos
Anopheles/parasitologia , Interações Hospedeiro-Parasita , Malária Falciparum/prevenção & controle , Doenças Mitocondriais/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Resistência à Doença , Feminino , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/parasitologia , Humanos , Proteínas de Insetos/biossíntese , Masculino , Mitocôndrias/metabolismo , Mitocôndrias/parasitologia , Mitocôndrias/ultraestrutura , Doenças Mitocondriais/parasitologia , Transdução de Sinais
3.
J Exp Biol ; 216(Pt 2): 208-17, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23255191

RESUMO

The highly conserved insulin/insulin-like growth factor (IGF) signaling (IIS) pathway regulates metabolism, development, lifespan and immunity across a wide range of organisms. Previous studies have shown that human insulin ingested in the blood meal can activate mosquito IIS, resulting in attenuated lifespan and increased malaria parasite infection. Because human IGF1 is present at higher concentrations in blood than insulin and is functionally linked with lifespan and immune processes, we predicted that human IGF1 ingested in a blood meal would affect lifespan and malaria parasite infection in the mosquito Anopheles stephensi. Here we demonstrate that physiological levels of ingested IGF1, like insulin, can persist intact in the blood-filled midgut for up to 30 h and disseminate into the mosquito body, and that both peptides activate IIS in mosquito cells and midgut. At these same levels, ingested IGF1 alone extended average mosquito lifespan by 23% compared with controls and, more significantly, when ingested in infected blood meals, reduced the prevalence of Plasmodium falciparum-infected mosquitoes by >20% and parasite load by 35-50% compared with controls. Thus, the effects of ingested IGF1 on mosquito lifespan and immunity are opposite to those of ingested insulin. These results offer the first evidence that insect cells can functionally discriminate between mammalian insulin and IGF1. Further, in light of previous success in genetically targeting IIS to alter mosquito lifespan and malaria parasite transmission, this study indicates that a more complete understanding of the IIS-activating ligands in blood can be used to optimize transgenic strategies for malaria control.


Assuntos
Anopheles/parasitologia , Interações Hospedeiro-Parasita , Insetos Vetores/parasitologia , Fator de Crescimento Insulin-Like I/metabolismo , Malária/transmissão , Plasmodium falciparum/patogenicidade , Animais , Anopheles/fisiologia , Linhagem Celular , Sistema Digestório/metabolismo , Sistema Digestório/parasitologia , Eritrócitos/parasitologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Proteínas de Insetos/metabolismo , Insetos Vetores/fisiologia , Insulina/metabolismo , Malária/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
4.
PLoS Pathog ; 6(8)2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20714345

RESUMO

Malaria (Plasmodium spp.) kills nearly one million people annually and this number will likely increase as drug and insecticide resistance reduces the effectiveness of current control strategies. The most important human malaria parasite, Plasmodium falciparum, undergoes a complex developmental cycle in the mosquito that takes approximately two weeks and begins with the invasion of the mosquito midgut. Here, we demonstrate that increased Akt signaling in the mosquito midgut disrupts parasite development and concurrently reduces the duration that mosquitoes are infective to humans. Specifically, we found that increased Akt signaling in the midgut of heterozygous Anopheles stephensi reduced the number of infected mosquitoes by 60-99%. Of those mosquitoes that were infected, we observed a 75-99% reduction in parasite load. In homozygous mosquitoes with increased Akt signaling parasite infection was completely blocked. The increase in midgut-specific Akt signaling also led to an 18-20% reduction in the average mosquito lifespan. Thus, activation of Akt signaling reduced the number of infected mosquitoes, the number of malaria parasites per infected mosquito, and the duration of mosquito infectivity.


Assuntos
Anopheles/parasitologia , Malária/parasitologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sistema Digestório/parasitologia , Interações Hospedeiro-Parasita , Humanos , Estágios do Ciclo de Vida , Prevalência , Transdução de Sinais
5.
PLoS Pathog ; 6(7): e1001003, 2010 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-20664791

RESUMO

Malaria (Plasmodium spp.) kills nearly one million people annually and this number will likely increase as drug and insecticide resistance reduces the effectiveness of current control strategies. The most important human malaria parasite, Plasmodium falciparum, undergoes a complex developmental cycle in the mosquito that takes approximately two weeks and begins with the invasion of the mosquito midgut. Here, we demonstrate that increased Akt signaling in the mosquito midgut disrupts parasite development and concurrently reduces the duration that mosquitoes are infective to humans. Specifically, we found that increased Akt signaling in the midgut of heterozygous Anopheles stephensi reduced the number of infected mosquitoes by 60-99%. Of those mosquitoes that were infected, we observed a 75-99% reduction in parasite load. In homozygous mosquitoes with increased Akt signaling parasite infection was completely blocked. The increase in midgut-specific Akt signaling also led to an 18-20% reduction in the average mosquito lifespan. Thus, activation of Akt signaling reduced the number of infected mosquitoes, the number of malaria parasites per infected mosquito, and the duration of mosquito infectivity.


Assuntos
Anopheles/parasitologia , Interações Hospedeiro-Parasita , Estágios do Ciclo de Vida , Malária/parasitologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sistema Digestório/parasitologia , Humanos , Prevalência , Transdução de Sinais
6.
Gen Comp Endocrinol ; 173(2): 303-12, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21703270

RESUMO

Insulin-like peptides (ILPs) regulate a multitude of biological processes, including metabolism and immunity to infection, and share similar structural motifs across widely divergent taxa. Insulin/insulin-like growth factor signaling (IIS) pathway elements are similarly conserved. We have shown that IIS regulates reproduction, innate immunity, and lifespan in female Anopheles stephensi, a major mosquito vector of human malaria. To further explore IIS regulation of these processes, we identified genes encoding five ILPs in this species and characterized their expression in tissues. Antisera to ILP homologs in Anopheles gambiae were used to identify cellular sources in An. stephensi females by immunocytochemistry. We analyzed tissue-specific ILP transcript expression in young and older females, in response to different feeding regimens, and in response to infection with Plasmodiumfalciparum with quantitative reverse transcriptase-PCR assays. While some ILP transcript changes were evident in older females and in response to blood feeding, significant changes were particularly notable in response to hormonal concentrations of ingested human insulin and to P. falciparum infection. These changes suggest that ILP secretion and action may be similarly responsive in Plasmodium-infected females and potentially alter metabolism and innate immunity.


Assuntos
Anopheles/metabolismo , Anopheles/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Somatomedinas/metabolismo , Animais , Anopheles/genética , Feminino , Imuno-Histoquímica , Insulina/genética , Insulina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somatomedinas/genética
7.
Trends Parasitol ; 24(8): 333-6, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18603475

RESUMO

Quantitative analyses of malaria parasite development are necessary to assess the efficacy of control measures. Such analyses in the mammalian host have been difficult to implement, lagging behind the use of antiparasitic drugs, vaccine development and transmission-blocking strategies. Even less is known about the genetic, environmental and other factors that impact sporogony in the mosquito host. Here, we summarize current knowledge and review a first attempt to model sporogonic development quantitatively.


Assuntos
Culicidae/parasitologia , Insetos Vetores/parasitologia , Plasmodium falciparum/fisiologia , Animais , Evolução Biológica , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Modelos Biológicos
8.
Parasit Vectors ; 8: 424, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26283222

RESUMO

BACKGROUND: Fruit flies and mammals protect themselves against infection by mounting immune and metabolic responses that must be balanced against the metabolic needs of the pathogens. In this context, p38 mitogen-activated protein kinase (MAPK)-dependent signaling is critical to regulating both innate immunity and metabolism during infection. Accordingly, we asked to what extent the Asian malaria mosquito Anopheles stephensi utilizes p38 MAPK signaling during infection with the human malaria parasite Plasmodium falciparum. METHODS: A. stephensi p38 MAPK (AsP38 MAPK) was identified and patterns of signaling in vitro and in vivo (midgut) were analyzed using phospho-specific antibodies and small molecule inhibitors. Functional effects of AsP38 MAPK inhibition were assessed using P. falciparum infection, quantitative real-time PCR, assays for reactive oxygen species and survivorship under oxidative stress, proteomics, and biochemical analyses. RESULTS: The genome of A. stephensi encodes a single p38 MAPK that is activated in the midgut in response to parasite infection. Inhibition of AsP38 MAPK signaling significantly reduced P. falciparum sporogonic development. This phenotype was associated with AsP38 MAPK regulation of mitochondrial physiology and stress responses in the midgut epithelium, a tissue critical for parasite development. Specifically, inhibition of AsP38 MAPK resulted in reduction in mosquito protein synthesis machinery, a shift in glucose metabolism, reduced mitochondrial metabolism, enhanced production of mitochondrial reactive oxygen species, induction of an array of anti-parasite effector genes, and decreased resistance to oxidative stress-mediated damage. Hence, P. falciparum-induced activation of AsP38 MAPK in the midgut facilitates parasite infection through a combination of reduced anti-parasite immune defenses and enhanced host protein synthesis and bioenergetics to minimize the impact of infection on the host and to maximize parasite survival, and ultimately, transmission. CONCLUSIONS: These observations suggest that, as in mammals, innate immunity and mitochondrial responses are integrated in mosquitoes and that AsP38 MAPK-dependent signaling facilitates mosquito survival during parasite infection, a fact that may attest to the relatively longer evolutionary relationship of these parasites with their invertebrate compared to their vertebrate hosts. On a practical level, improved understanding of the balances and trade-offs between resistance and metabolism could be leveraged to generate fit, resistant mosquitoes for malaria control.


Assuntos
Anopheles/imunologia , Metabolismo Energético , Imunidade Inata , Plasmodium falciparum/imunologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Anopheles/metabolismo , Perfilação da Expressão Gênica , Estresse Oxidativo , Proteoma/análise , Espécies Reativas de Oxigênio/análise , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sobrevida
9.
Microbes Infect ; 15(12): 775-87, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23774695

RESUMO

The insulin/insulin-like growth factor signaling (IIS) cascade is highly conserved and regulates diverse physiological processes such as metabolism, lifespan, reproduction and immunity. Transgenic overexpression of Akt, a critical regulator of IIS, was previously shown to shorten mosquito lifespan and increase resistance to the human malaria parasite Plasmodium falciparum. To further understand how IIS controls mosquito physiology and resistance to malaria parasite infection, we overexpressed an inhibitor of IIS, phosphatase and tensin homolog (PTEN), in the Anopheles stephensi midgut. PTEN overexpression inhibited phosphorylation of the IIS protein FOXO, an expected target for PTEN, in the midgut of A. stephensi. Further, PTEN overexpression extended mosquito lifespan and increased resistance to P. falciparum development. The reduction in parasite development did not appear to be due to alterations in an innate immune response, but rather was associated with increased expression of genes regulating autophagy and stem cell maintenance in the midgut and with enhanced midgut barrier integrity. In light of previous success in genetically targeting the IIS pathway to alter mosquito lifespan and malaria parasite transmission, these data confirm that multiple strategies to genetically manipulate IIS can be leveraged to generate fit, resistant mosquitoes for malaria control.


Assuntos
Anopheles/imunologia , Expressão Gênica , Interações Hospedeiro-Patógeno , PTEN Fosfo-Hidrolase/biossíntese , Plasmodium falciparum/crescimento & desenvolvimento , Animais , Anopheles/enzimologia , Anopheles/genética , Anopheles/parasitologia , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/metabolismo , Autofagia , Feminino , Longevidade , PTEN Fosfo-Hidrolase/genética , Plasmodium falciparum/imunologia , Células-Tronco/fisiologia
10.
Cell Tissue Res ; 329(2): 391-408, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17450383

RESUMO

The crustacean hyperglycemic hormone (CHH) peptides regulate diverse physiological processes from reproduction to metabolism and molting in arthropods. In insects, the ion transport peptides (ITP), also members of the CHH family, have only been implicated in ion transport. In this study, we sequenced a nucleotide fragment spanning the conserved A1/A2 region of the putative CHH/ITP gene. This fragment was amplified from larval cDNA of the tobacco hornworm, Manduca sexta and showed a high degree of sequence conservation with the same region from other insects and, to a lesser degree, with that of crustacean species, suggesting the presence of a Manduca-specific CHH/ITP mRNA (MasITP mRNA). CHH-like immunocytochemical analyses with two crustacean antisera (from Carcinus maenas and Cancer pagurus) identified the presence of CHH-like immunoreactivity in nervous tissue of all developmental stages, but not in the gut of M. sexta. Specifically, CHH-like peptides localized to paired type IA(2) neurosecretory cells of the pars lateralis of the brain (projecting ipsilaterallly to the corpora cardiaca-allata complex) and to neurosecretory cells and transverse nerves of the ventral nerve cord in larvae, pupae, and adults. The distribution of the putative MasITP peptide shifted during development in a manner consistent with metamorphic reorganization. A comparison of hemolymph equivalents of CHH detected by enzyme-linked immunosorbent assay with CHH-like immunoreactivity in transverse nerves provided evidence for the release of MasITP from the transverse nerves into the hemolymph at insect ecdysis. These data suggest the presence of an insect ITP in M. sexta and a role for this hormone during ecdysis.


Assuntos
Hormônios de Invertebrado/metabolismo , Manduca/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes , Sequência de Bases , Sistema Nervoso Central/metabolismo , Sequência Conservada , Hemolinfa/metabolismo , Imuno-Histoquímica , Hormônios de Invertebrado/sangue , Transporte de Íons , Larva/metabolismo , Manduca/crescimento & desenvolvimento , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/sangue , Radioimunoensaio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA