The complex interplay between mosquito positive and negative regulators of Plasmodium development

https://doi.org/10.1016/j.mib.2005.06.013Get rights and content

The malaria parasite, Plasmodium, requires sexual development in the mosquito before it can be transmitted to the vertebrate host. Mosquito genes are able to substantially modulate this process, which can result in major decreases in parasite numbers. Even in susceptible mosquitoes, haemolymph proteins implicated in systemic immune reactions, together with local epithelial responses, cause lysis of more than 80% of the ookinetes that cross the mosquito midgut. In a refractory mosquito strain, immune responses lead to melanisation of virtually all parasites. Conversely, certain mosquito genes have an opposite effect: they are used by the parasite to evade defence reactions. Detailed understanding of the interplay between positive and negative regulators of parasite development could lead to the generation of novel approaches for malaria control through the vector.

Introduction

Malaria is a leading infectious disease that is responsible for up to three million deaths annually, mainly among young children in Sub-Saharan Africa. It is caused by the protozoan parasite Plasmodium, which requires Anopheles mosquitoes for transmission as it is in these that the parasite undergoes sexual development. However, of the several hundred Anopheline species only around 70 can transmit human malaria, and of these only half are significant vectors [1]. For example, in the Anopheles funestus species complex only two of the nine recognized species are important vectors. Vectorial capacity can also vary widely between and within different populations or strains of the same mosquito species [2, 3], which suggests the existence of genetic refractory mechanisms that might help to design new strategies for malaria control.

Anopheles gambiae is the main vector of human malaria in Africa. This has become a workable model through the development of an important toolkit that includes: genetic and chromosome maps [4, 5], transgenic technology [6], a method for reverse genetic analysis by double-stranded RNA-mediated RNAi in cell lines and in adult mosquitoes [7, 8], a set of cultured cell lines [9], an extensive collection of cDNAs and expressed sequence tag sequences [10, 11], DNA microarrays [12, 13], and recently — and most importantly — a full genome sequence [14] and the related bioinformatics resources [15, 16]. These tools, in combination with the development of molecular markers and recent advances in image analysis, have been vital for deciphering the cellular and molecular mechanisms that underlie vector–parasite interactions.

Below, we review the current understanding of mosquito genes that affect and, in many cases, that determine Plasmodium development in the vector. A key concept that emerged from recent studies is the existence of a fine balance between mosquito factors that negatively or positively affect the development of the parasite [17, 18••]. To date, such factors have been shown to act during penetration of the mosquito midgut by Plasmodium (Figure 1). This step is the strongest bottleneck of the malaria transmission cycle in the vector, and has been studied extensively. Furthermore, other genetically selected strains have revealed that refractory mechanisms operate during or immediately after midgut invasion [2, 19]. For these reasons, the subsequent sections of the review are organised relative to the invasion process.

Section snippets

Vector–parasite interactions before midgut invasion

The first step towards malaria transmission is the ingestion of Plasmodium gametocytes by female mosquitoes during a bloodmeal. In the midgut lumen, gametocytes rapidly differentiate into male and female gametes, which then fuse to produce zygotes (Figure 1). In some mosquito species, male gametogenesis (a process known as exflagellation) has never been recorded, which suggests either the absence of activators or the presence of inhibitors. An important factor that is required for induction of

Vector–parasite interactions during midgut invasion

The great majority of parasites are lost during midgut invasion — the most crucial step in the sexual development of a parasite [23]. However, the presence of just a few surviving ookinetes is sufficient to establish successful transmission: soon after invasion they transform into oocysts that undergo serial mitotic divisions, with each one producing several thousand sporozoites. It remains unknown whether or not midgut invasion is receptor-mediated. Recently, annexins (which are strongly

Vector–parasite interactions after midgut invasion

Surviving ookinetes transform into oocysts after they emerge at the basal subepithelial space of the midgut (Figure 1). At least two components of the midgut basal lamina — laminin and collagen — are implicated in this transition [23]. Several ookinete surface and secreted proteins, including P28, P25, CTRP and SOAP, are known to bind to these two molecules [49, 50, 51], however, the role of these interactions remains unknown. To date, the minimal set of components from both the vector and the

Conclusions

Our understanding of the molecular basis of vector–parasite interactions, especially of the model P. bergheiA. gambiae system, has greatly advanced during the past five years. It is now well-established that the mosquito immune system is activated during infection, and that immune genes and reactions account for substantial parasite losses, especially during invasion of the mosquito midgut (even though the immune system often allows immune escape and parasite transmission). Additional classes

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We thank GK Christophides for fruitful discussions and T Schlegelmilch for helping in manuscript presentation. The work of our laboratory discussed in this review was supported by a National Institute of Allergy and Infectious Diseases–National Institutes of Health Programme Project grant 2PO1AI044220-06A1, a European Commission Research Training Network grant HPRN-CT-2000-00080, a European Commission Network of Excellence grant (BioMalPar) and the European Molecular Biology Laboratory (EMBL).

References (53)

  • D. Vlachou et al.

    Anopheles gambiae laminin interacts with the P25 surface protein of Plasmodium berghei ookinetes

    Mol Biochem Parasitol

    (2001)
  • F.H. Collins et al.

    Genetic selection of a Plasmodium-refractory strain of the malaria vector Anopheles gambiae

    Science

    (1986)
  • O. Niare et al.

    Genetic loci affecting resistance to human malaria parasites in a West African mosquito vector population

    Science

    (2002)
  • M. Coluzzi et al.

    A polytene chromosome analysis of the Anopheles gambiae species complex

    Science

    (2002)
  • L. Zheng et al.

    An integrated genetic map of the African human malaria vector mosquito, Anopheles gambiae

    Genetics

    (1996)
  • G.L. Grossman et al.

    Germline transformation of the malaria vector, Anopheles gambiae, with the piggyBac transposable element

    Insect Mol Biol

    (2001)
  • S. Blandin et al.

    Reverse genetics in the mosquito Anopheles gambiae: targeted disruption of the Defensin gene

    EMBO Rep

    (2002)
  • G. Dimopoulos et al.

    Anopheles gambiae pilot gene discovery project: identification of mosquito innate immunity genes from expressed sequence tags generated from immune-competent cell lines

    Proc Natl Acad Sci USA

    (2000)
  • S.M. Gomez et al.

    Pilot Anopheles gambiae full-length cDNA study: sequencing and initial characterization of 35,575 clones

    Genome Biol

    (2005)
  • A.N. Dana et al.

    Gene expression patterns associated with blood-feeding in the malaria mosquito Anopheles gambiae

    BMC Genomics

    (2005)
  • G. Dimopoulos et al.

    Genome expression analysis of Anopheles gambiae: responses to injury, bacterial challenge, and malaria infection

    Proc Natl Acad Sci USA

    (2002)
  • R.A. Holt et al.

    The genome sequence of the malaria mosquito Anopheles gambiae

    Science

    (2002)
  • E.V. Kriventseva et al.

    AnoEST: Toward A. gambiae functional genomics

    Genome Res

    (2005)
  • M.A. Osta et al.

    Effects of mosquito genes on Plasmodium development

    Science

    (2004)
  • K.D. Vernick et al.

    Plasmodium gallinaceum: a refractory mechanism of ookinete killing in the mosquito, Anopheles gambiae

    Exp Parasitol

    (1995)
  • O. Billker et al.

    Calcium and a calcium-dependent protein kinase regulate gamete formation and mosquito transmission in a malaria parasite

    Cell

    (2004)
  • Cited by (26)

    • Time- and tissue-specific antimicrobial activity of the common bed bug in response to blood feeding and immune activation by bacterial injection

      2021, Journal of Insect Physiology
      Citation Excerpt :

      However, many common immune pathway components, including STAT, Spatzle, and Drosha, are not upregulated in bed bugs in response to Gram-negative bacteria (Potts et al., 2020). Biological transmission of parasites and pathogens by vectors is regulated by a complex interplay between host and parasite (e.g., Castillo et al., 2011; Marliére et al., 2015; Saporito et al., 2013; Vlachou & Kafatos, 2005). Inside the vectors, some organisms replicate, replicate and change developmental form, or just change developmental stages (Childs & Prosper, 2020; Cordero-Montoya et al., 2019; Dostálová & Volf, 2012; Kollien & Schaub, 2000; Sacks & Kamhawi, 2001).

    • Ecological immunology of mosquito-malaria interactions

      2008, Trends in Parasitology
      Citation Excerpt :

      In Aedes aegypti, selection for early and late pupation resulted in correlated changes in body size and melanization response, revealing trade-offs between those traits [31]. In Anophelines, microarray studies have shown that the invasion of the mosquito midgut and salivary glands by Plasmodium parasites induces the transcription of several immune-related molecules [3] (reviewed in [32–34]). The production of these molecules is expected to be energetically costly and to divert resources away from growth and maintenance [1].

    • Chapter 1 Strain Theory of Malaria. The First 50 Years

      2008, Advances in Parasitology
      Citation Excerpt :

      High gametocyte production in response to sulphadoxine—pyrimethamine suggests that gametocyte production can respond to some cues within the host, and is not a fixed trait (Barnes and White, 2005). Incompatibility between parasite strains and various species and strains of Anopheles is now believed to reflect an interplay between polymorphic innate immune response genes of the mosquito that promote or retard parasite killing (melanization) and unknown melanization resistance genes of the parasite (Vlachou and Kafatos, 2005), hence current discussions often invoke evolutionary costs (Lambrechts et al., 2005; Sinden et al., 2004). Simultaneous propagation of multiple strains of malaria by a single mosquito has been confirmed by molecular genetic studies (Babiker et al., 1994; Huber et al., 1998; Ranford-Cartwright et al., 1993), but the high frequency of recombination during the sexual phase of the parasite life-cycle (confirming that ‘male gametocytes of one strain’ can indeed ‘fertilize the females of a different strain’) may mean that outcrossing, between strains, is as common as ‘selfing’ and thus, as predicted by Shute and Maryon (1954), that strains rapidly lose their identity in endemic areas.

    • The developmental migration of Plasmodium in mosquitoes

      2006, Current Opinion in Genetics and Development
    View all citing articles on Scopus
    1

    Current address: Department of Biological Sciences, Imperial College London, SW7 2AZ, London, UK

    View full text