Skip to main content
Erschienen in: Malaria Journal 1/2020

Open Access 01.12.2020 | Research

Comparative assessment of insecticide resistance phenotypes in two major malaria vectors, Anopheles funestus and Anopheles arabiensis in south-eastern Tanzania

verfasst von: Polius G. Pinda, Claudia Eichenberger, Halfan S. Ngowo, Dickson S. Msaky, Said Abbasi, Japhet Kihonda, Hamis Bwanaly, Fredros O. Okumu

Erschienen in: Malaria Journal | Ausgabe 1/2020

Abstract

Background

Long-lasting insecticide-treated nets (LLINs) and indoor residual spraying (IRS) have greatly reduced malaria transmission in sub-Saharan Africa, but are threatened by insecticide resistance. In south-eastern Tanzania, pyrethroid-resistant Anopheles funestus are now implicated in > 80% of malaria infections, even in villages where the species occurs at lower densities than the other vector, Anopheles arabiensis. This study compared the insecticide resistance phenotypes between the two malaria vectors in an area where pyrethroid-LLINs are widely used.

Methods

The study used the World Health Organization (WHO) assays with 1×, 5× and 10× insecticide doses to assess levels of resistance, followed by synergist bioassays to understand possible mechanisms of the observed resistance phenotypes. The tests involved adult mosquitoes collected from three villages across two districts in south-eastern Tanzania and included four insecticide classes.

Findings

At baseline doses (1×), both species were resistant to the two candidate pyrethroids (permethrin and deltamethrin), but susceptible to the organophosphate (pirimiphos-methyl). Anopheles funestus, but not An. arabiensis was also resistant to the carbamate (bendiocarb). Both species were resistant to DDT in all villages except in one village where An. arabiensis was susceptible. Anopheles funestus showed strong resistance to pyrethroids, surviving the 5× and 10× doses, while An. arabiensis reverted to susceptibility at the 5× dose. Pre-exposure to the synergist, piperonyl butoxide (PBO), enhanced the potency of the pyrethroids against both species and resulted in full susceptibility of An. arabiensis (> 98% mortality). However, for An. funestus from two villages, permethrin-associated mortalities after pre-exposure to PBO only exceeded 90% but not 98%.

Conclusions

In south-eastern Tanzania, where An. funestus dominates malaria transmission, the species also has much stronger resistance to pyrethroids than its counterpart, An. arabiensis, and can survive more classes of insecticides. The pyrethroid resistance in both species appears to be mostly metabolic and may be partially addressed using synergists, e.g. PBO. These findings may explain the continued persistence and dominance of An. funestus despite widespread use of pyrethroid-treated LLINs, and inform new intervention choices for such settings. In short and medium-term, these may include PBO-based LLINs or improved IRS with compounds to which the vectors are still susceptible.
Hinweise
Polius G. Pinda and Claudia Eichenberger contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ITS2
Internal transcribed spacer 2 region
IGS
Intergenic spacer regions
IHI
Ifakara Health Institute
IRB
Institutional Review Board
LLINs
Long-lasting insecticidal nets
NIMR
National Institute for Medical Research
qPCR
Quantitative polymerase chain reaction
CDC
Centre for disease control and prevention
GSTs
Glutathione-S-transferase enzymes

Background

Effective use of long-lasting insecticide-treated nets (LLINs) and indoor residual spraying (IRS) has tremendously reduced malaria transmission in sub-Saharan Africa [1, 2]. Despite this reduction, malaria transmission continues in several areas, driven by mosquitoes that are either physiologically [35] or behaviourally resistant [610] to current insecticide-based interventions. Resistance to commonly used pyrethroids has also necessitated a change of insecticide classes for IRS to either carbamates, organophosphates or more recently neonicotinoids approved by the World Health Organization (WHO) [11]. Similarly new insecticide-treated nets (ITNs) are being developed that contain either multiple insecticide classes [12] or pyrethroids and synergists [13, 14], and are expected to improve the control of resistant mosquitoes.
Mosquito resistance involves different mechanisms, through which they can withstand exposures to insecticides. These include metabolic resistance, target-site resistance, behavioural resistance, and cuticular resistance [1517]. The discriminating concentration is used to evaluate the phenotypic resistance which when detected, the level of resistance can be subsequentlly determined by using intensity assays. The intensity assays uses the 5× and 10× discriminating concentration in a stepwise manner aiming at providing the information on the range of resistance present in a target vector [18]. Mosquitoes that express metabolic forms of resistance produce large quantities of enzymes or alternate the enzyme catalytic centre to efficiently detoxify the insecticide. The specific enzymes include monooxygenases (i.e. cytochrome P450s), which detoxify pyrethroids and carbamates, glutathione-S-transferases (GSTs), which detoxify organochlorides like DDT [17], and esterases, which detoxify pyrethroids and organophosphates [19, 20]. The degree to which the enzymatic proteins are expressed, and the level of resistance can be assessed using quantitative polymerase chain reaction (qPCR). Phenotypic assays use synergists, such as piperonyl butoxide (PBO), which enhance the potency of an insecticide by inhibiting the enzymes responsible for the insecticide metabolism [21]. On the other hand, some mosquitoes may have one or multiple target-site mutations due to modification of protein receptors usually targeted by insecticides (e.g. the voltage-gated sodium channels targeted by pyrethroids and organochlorides), thereby blocking or reducing the effectiveness of the insecticides [2224]. Recently, scientists have also demonstrated that a sensory appendage protein (SAP2) enriched in the legs of malaria-carrying mosquitoes can also confer resistance to insecticides, thus allowing these mosquitoes to survive contact with ITNs [25].
Different Anopheles species have diverse levels of competencies in pathogen transmission, and also respond different to interventions based on their behaviour and physiology [26, 27]. With the rise of insecticide resistance in the vector populations, the choice of interventions will depend on characteristics of the local vectors. Comprehensive understanding of the distribution and underlying mechanisms of insecticide resistance is, therefore, important for planning and implementing vector control interventions.
In the villages of south-eastern Tanzania, where Anopheles funestus have been reported to be implicated in most of the ongoing malaria transmission [2830], signs of resistance to most public health pesticides have been observed. Anopheles funestus also appears to survive longer than other co-existing vector species (parity rates are higher than Anopheles arabiensis) [28]. However, since the species is highly anthropophagic (prefers to blood-feed on humans over that of other vertebrates) [31, 32] and endophilic (prefers to bite indoors) [33], one would expect its populations and transmission activity to have been significantly reduced by ITNs now widely used in Tanzania for more than one decade [3437]. Indeed, historical evidence from both East and Southern Africa suggests that effective insecticide-based indoor interventions can eliminate An. funestus on a local scale [38, 39]. Anopheles gambiae sensu stricto (s.s.), which is generally considered the most competent malaria vector, shares similar behaviours with An. funestus, i.e. high degree of anthropophily and endophilly [31, 32, 40]. However, An. gambiae s.s. unlike An. funestus has been highly impacted by ITNs in Kenya and Tanzania [4143].
An important question, therefore, is why and how An. funestus, despite being highly anthropophagic and endophilic, survived the ITN onslaught, and why it continues to mediate most malaria transmission in rural south-eastern Tanzania despite co-occurrence with a different malaria vector species, An. arabiensis. One hypothesis has been that An. funestus expresses higher intensities of resistance to most of the commonly used insecticides in comparison to other malaria vectors and is, therefore, far less impacted by insecticidal interventions. A meta-analysis of various datasets has verified this phenomenon at global scale [44], but specific field tests comparing resistance intensities in the different vector species are limited. As shown by Kaindoa et al. in a study conducted in south-eastern Tanzania, An. funestus was resistant to pyrethroids, organochlorides, and carbamates [28]. Another study from the same study area demonstrated that resistance of An. arabiensis to diagnostic insecticide concentrations varied between nearby locations and seasons [45]. However, the intensity and mechanisms of these resistance phenotypes were not compared between species.
This study, therefore, compared the intensities and knockdown time of insecticide-resistance between the two main malaria vectors, An. funestus and An. arabiensis, in rural south-eastern Tanzania. Potential involvement of metabolic resistance and insecticide potency enhancement by Piperonyl Butoxide (PBO) synergists was also assessed.

Methods

Study site

Mosquito collections were done in three different villages, namely; Ikwambi (7.98033°S, 36.81701°E) and Sululu (8.00324°S, 36.83118°E) in Kilombero district, and Tulizamoyo village (8.35747°S, 36.70664°E) in Ulanga district, south-eastern Tanzania (Fig. 1). The main malaria vectors in this area include An. arabiensis and An. funestus, with the latter driving more than 80% of the malaria transmission [28, 29]. This area has had high coverage of pyrethroid-treated nets for several years, but no IRS is implemented. The villages are all in low altitude areas, rising not more than 500 m above sea level. Mean daily temperatures are 20–33 °C, annual rainfall, 1200–1800 mm and relative humidity ranged between 24 and 97% [46, 47]. Most community members here are farmers, cultivating rice, maize and other crops in the Kilombero river valley.

Mosquito collection

The WHO protocol for insecticide susceptibility tests [18] was used with slight modifications to conduct the basic bioassays and the resistance intensity assays. Since An. funestus mosquitoes were difficult to find as larvae across all the study villages, young nulliparous adult females of both An. funestus and An. arabiensis were used instead of larval collections. Mosquitoes were collected from September 2018 to November 2019 using CDC light traps [48]. Collections were done from 07.00 p.m. to 07.00 a.m. each night. To maximize probabilities of getting young unfed nulliparous females, houses near the edges of the villages and near potential habitats were selected for collections, based on previously-described heterogeneity of malaria transmission [49].
The traps were hung beside human occupied bed nets [50], but with extended catch bags to improve survival of the mosquitoes for subsequent assays. Each morning, after collections, the mosquitoes were transported to the Ifakara Health Institute’s mosquito biology laboratory, VectorSphere, in Ifakara and maintained at 27 ± 2 °C and 80 ± 10% relative humidity for 24 h to acclimatize as previously described [45]. During the acclimatization period, mosquitoes were supplied with 10% glucose solution. The mosquitoes were identified morphologically using the Gillies and Coetzee identification key [51], and non-target species were discarded. Tests were conducted using only non-blood-fed An. funestus and An. arabiensis females.

Bioassays

This study involved three steps: (i) susceptibility assays to examine phonotypic resistance against standard insecticide doses, (ii) tests to assess intensity of resistance against chemicals to which resistance had been detected at baseline level, and (iii) tests using synergists to assess possible mechanisms of the observed reistance.
Baseline insecticide susceptibility bioassays were done according to WHO guidelines [18]. Candidate insecticides were selected from four classes as follows: organophosphate (0.25% pirimiphos-methyl), organochloride (4% DDT), carbamate (0.1% bendiocarb), pyrethroid type I (0.75%, 3.75 and 7.5% permethrin) and pyrethroid type II (0.05%, 0.25 and 0.5% deltamethrin). In each test, 120 mosquitoes were exposed to the insecticide-impregnated papers, and oil-impregnated papers as controls. Each test comprised six replicates (four treatments and two controls) with the total of 120 mosquitoes. Mosquitoes were exposed for 1 h and the knockdown time recorded at an interval of 10, 15, 20, 30, 40, 50, 60 min. They were then transferred to holding tubes, provided with 10% glucose solution, and their mortality recorded after 24 h.
Where resistance was observed in the baseline assays with standard diagnostic doses (i.e. 1×), additional tests were done to assess intensities of the resistance using 5× and 10× multiplicative doses of the insecticides. These included tests against 3.75% and 7.5% permethrin, and 0.25% and 0.5% deltamethrin. The procedures were similar to the baseline tests to assess the mortality.
Lastly, 4% Piperonyl Butoxide (PBO), a synergist, was used to assess the possible resistance mechanism by attempting to reverse the observed mortality outcomes [18]. Each test had four groups, each with 80 mosquitoes (in groups of 20), treated as follows: the first cohort was exposed to 4% PBO for one hour and immediately exposed to deltamethrin or permethrin for 60 min, a second group was exposed directly to the respective insecticides (i.e. deltamethrin, permethrin), a third group was exposed to the PBO only and the fourth group was exposed to control papers impregnated by silicone oil but no insecticide nor synergist. Given test kit limitations, the PBO tests were done only for pyrethroids.

Molecular identification of sibling species of the tested mosquitoes

Up to 10% of the mosquitoes from each bioassay were packed separately and labelled with information about experimental date, village name, type of insecticide, insecticide dose used, species of mosquito, replicate number and sample ID. The packed mosquitoes were sent to the laboratory for molecular species identification of sibling species in the An. funestus and An. gambiae s.l. complexes, using DNA extracted from the mosquito legs. Polymerase chain reaction assays were conducted based on species-specific nucleotide sequences of the ribosomal DNA (rDNA) by relying on the intergenic spacer regions (IGS) for An. gambiae sensu lato (s.l.) members and the non-coding internal transcribed spacer 2 region (ITS2) for An. funestus [52, 53]. DNA bands were photographed under ultraviolet light using Kodak Gel Logic 100 imaging system [54].

Data analysis

The data on insecticide susceptibility was interpreted based on the WHO-specified thresholds for resistance determination [18]. Susceptibility was confirmed when mortality was ≥ 98%, possible resistance was determined when mortality ranged from 90 to 97%, in which case the tests were repeated for confirmation, and resistance was confirmed when mortality was < 90%. When mortality greater than 10% was observed in controls, the test mortality was corrected using Abbott’s formula to avoid the biased estimations [55]. Tests were discarded and repeated, whenever control mortality exceeded 20% [18]. Final results were plotted in graphs using R software version 3.0 [56]. Log-probity analysis was used to calculate mean duration at which 50% (KDT50) and 95% (KDT95) of mosquitoes exposed to specific insecticides were knocked down.

Results

Phenotypic resistance at baseline insecticide concentrations

Both species were resistant to the pyrethroids (permethrin and deltamethrin) and the organochloride (DDT), but susceptible to the organophosphate (pirimiphos-methyl) at standard baseline doses (1×). There was general susceptibility to the carbamate (bendiocarb) by both species across the study area, except in one of the villages, Tulizamoyo, where An. funestus were resistant to this insecticide. Anopheles funestus generally showed lower mortalities to the insecticides in the baseline tests compared to An. arabiensis (Fig. 2).

Phenotypic resistance at 5 and 10 times baseline concentrations

Anopheles funestus populations from Ikwambi and Tulizamoyo are resistant to both 5× and 10× concentrations of permethrin, but the same species from Sululu were susceptible to 10× permethrin concentrations (Fig. 3). For An. arabiensis on the other hand, resistance intensity declined with increasing insecticide concentrations. Their resistance to pyrethroids was already overcome at 5× doses in Ikwambi and Tulizamoyo villages, while the ones from Sululu village, which survived 5× doses, were overcome at 10× doses. At 10× doses, An. arabiensis from all the villages were completely susceptible to the two pyrethroids (Fig. 3). Because of the observed susceptibilities at baseline doses (Fig. 1), no intensity assays were done against pirimiphos-methyl or bendiocarb on either of the species.

Knockdown times (KDT)

Knockdown time for both mosquito species varied between insecticide and study villages. Consistently, the knockdown time slowed as the concentration increased for pyrethroid-exposed mosquitoes. Prolonged KDT95 (51–245 min) was observed to An. funestus exposed to pyrethroids compared to An. arabiensis (16–76 min), similar trend was observed on KDT50. Furthermore, the lowest knockdown time was observed when the mosquitoes were exposed to bendiocarb both at KDT50 and KDT95 for An. funestus and An. arabiensis (Table 1).
Table 1
The knockdown times of An. funestus and An. arabiensis mosquitoes at different insecticide concentrations
Insecticide
Village
Concentration
An. arabiensis
An. funestus
(Fold)
KDT50 ± SE (min)
KDT95 ± SE (min)
KDT50 ± SE (min)
KDT95 ± SE (min)
Deltamethrin
Ikwambi
40.87 ± 12.55
76.68 ± 30.88
77.35 ± 50.19
128.13 ± 111.12
8.65 ± 6.70
23.34 ± 11.67
59.03 ± 22.43
99.19 ± 54.25
10×
39.06 ± 12.85
77.21 ± 32.73
Sululu
38.81 ± 10.05
66.76 ± 21.76
86.39 ± 71.94
132.00 ± 146.02
11.61 ± 13.10
47.08 ± 23.83
68.15 ± 39.38
124.38 ± 97.13
10×
8.63 ± 8.84
29.22 ± 15.50
45.94 ± 15.73
87.10 ± 41.42
Tulizamoyo
31.30 ± 8.82
56.48 ± 18.01
152.37 ± 388.44
245.22 ± 702.53
16.16 ± 5.75
29.99 ± 12.05
50.54 ± 19.60
96.49 ± 52.42
10×
52.53 ± 16.60
88.72 ± 40.44
Permethrin
Ikwambi
42.19 ± 10.82
71.25 ± 24.19
104.11 ± 124.69
177.35 ± 257.04
6.97 ± 5.17
16.15 ± 7.61
37.62 ± 11.07
70.12 ± 25.75
10×
33.21 ± 11.87
71.14 ± 29.92
Sululu
25.35 ± 9.05
52.67 ± 19.02
82.76 ± 61.03
130.09 ± 127.92
4.05 ± 11.16
25.13 ± 15.64
33.26 ± 10.01
63.31 ± 22.11
10×
7.83 ± 5.64
18.93 ± 8.92
20.21 ± 10.21
51.82 ± 21.66
Tulizamoyo
42.80 ± 11.56
73.95 ± 26.67
70.76 ± 41.28
124.31 ± 97.84
6.98 ± 8.14
24.26 ± 13.34
38.32 ± 13.87
80.67 ± 37.66
10×
29.16 ± 8.61
53.82 ± 17.50
Pirimiphos-methyl
Ikwambi
 
49.06 ± 9.51
70.40 ± 20.85
32.31 ± 6.62
46.15 ± 11.89
Sululu
57.12 ± 10.92
75.62 ± 26.77
48.30 ± 7.54
64.09 ± 15.25
Tulizamoyo
 
53.08 ± 14.22
83.01 ± 33.48
34.03 ± 4.70
40.42 ± 7.92
DDT
Ikwambi
 
44.82 ± 9.38
67.70 ± 19.65
97.77 ± 116.62
146.48 ± 222.68
Sululu
55.00 ± 11.41
76.66 ± 27.24
72.33 ± 37.37
119.73 ± 82.09
Tulizamoyo
 
61.06 ± 23.07
99.04 ± 54.65
69.53 ± 35.24
113.72 ± 80.51
Bendiocarb
Ikwambi
 
14.86 ± 4.43
24.27 ± 9.45
26.32 ± 6.75
42.21 ± 13.04
Sululu
19.29 ± 4.37
27.23 ± 9.40
25.18 ± 4.92
33.00 ± 8.65
Tulizamoyo
 
16.74 ± 3.36
22.85 ± 7.14
40.42 ± 7.61
57.80 ± 14.09
SE standard error, KDT50 time taken for 50% of the tested mosquitoes to be knocked-down, KDT95 time taken for 95% of the tested mosquitoes to be knocked-down

Effects of pre-exposure to the synergist, PBO

Pre-exposure to the synergist, PBO, significantly reversed the pyrethroid resistance in both An. arabiensis and An. funestus. The PBO assays achieved mortalities > 98% in most cases, except for An. funestus populations from Sululu and Tulizamoyo villages, for which permethrin-associated mortalities were reversed past 95%, but not 98%. The synergist assays on An. arabiensis from all study areas demonstrated highest restoration of susceptibility (Fig. 4).

Molecular identification of species

After the bioassays, a total of 305 An. funestus and 144 An. arabiensis were sent to the laboratory for sibling species identification. Of all the An. funestus assessed, successful PCR amplification was 76% (n = 233). Of those that amplified, 99% were An. funestus s.s. (n = 232), while one was amplified as Anopheles leesoni. For An. gambiae s.l. successful amplification was 92% (n = 132), all of which were identified as An. arabiensis. The rest did not amplify in the PCR assays (n = 12).

Discussion

In this study, both An. arabiensis and An. funestus were resistant to pyrethroids and DDT. However, An. funestus exhibited far lower mortalities when subjected to pyrethroids at either the baseline concentration, five times concentration or the ten times concentration in the intensity bioassays. This suggests that while An. funestus is strongly resistant to the pyrethroids, the level of resistance in An. arabiensis was either low or moderate. This is the first study to directly compare resistance intensities of these two vectors in the area, and therefore provides important information on potential performance of current or future interventions against malaria. Given the differential contribution of the two vectors to overall transmission, their responsiveness to insecticidal interventions is an important factor for consideration in the elimination efforts.
Initial findings from standard WHO susceptibility assays by Kaindoa et al. [28] and Matowo et al. [45] on the two malaria vectors in the same study area observed that the baseline mortalities were higher in An. arabiensis than An. funestus. This was the initial indication that the intensity of resistance would be different between the two species, and necessitated additional tests according to standard WHO assays [18]. The duration at which either 50% or 95% of mosquitoes would be knocked-down varied between species, insecticides and study villages (Table 1).
Knock-down time was high when mosquitoes were exposed to standard concentrations of pyrethroids, but slowed as the concentrations increased. Since no kdr mutation tests were done, it is not possible to determine whether these observations were associated with the voltage-gated sodium channel protein mutation [57]. The slowest knockdown time was observed when the mosquito species were exposed to bendiocarb, these findings still support the potency of the insecticide (Table 1). The new findings clearly demonstrate that An. funestus populations, despite being the more dominant vector of malaria in the area, would be much more difficult to control using current pyrethroid-based interventions, in particular the LLINs.
As demonstrated by Matowo et al. for both An. arabiensis and Culex mosquitoes [45, 58], there were signs of fine-scale spatial variations in insecticide resistance. For example, An. funestus populations from Tulizamoyo were resistant to bendiocarb but populations of the same species from the other two villages were susceptible to the same chemical (Fig. 2). Similarly, the mortality percentages observed at 5× and 10× doses varied between the villages (Figs. 2, 3). This might be attributed to differences in the use of agricultural pesticides for crop protection in these villages [59]. Surprisingly, An. arabiensis from Ikwambi were 100% susceptible to DDT, against which both species from the other study villages were resistant (Fig. 2), which further suggests fine-scale spatial differences in resistance profiles.
The currently observed dominance of An. funestus is likely to be contributed by their well-documented resistance to commonly used insecticides [28, 6064], their high survival probabilities in the wild [28, 30] and high levels of anthropophily [28, 31, 33]. Its dominance in areas where insecticidal interventions such as ITNs are widely implemented is particularly surprising given that scale-up of ITNs has coincided with significant declines in populations of other anthropophilic vectors such as An. gambiae s.s. [4143]. Today, in rural south-eastern Tanzania, An. funestus co-exists with other Anopheles species, namely An. arabiensis, An. leesoni, Anopheles coustani, Anopheles squamosus, Anopheles rivulorum and Anopheles pharoensis [28]. However, it is known to carry most of the ongoing malaria transmission, sometime implicating to nearly nine in every ten new cases, even in areas where it occurs in lower densities than An. arabiensis [28, 29]. It was thus hypothesized that its dominance may at least be partly driven by stronger insecticide resistance levels to insecticides commonly used for public health, notably the pyrethroids used on bed nets.
As insecticide resistance increases across Africa, some populations have been observed to withstand up to 1000 times the standard concentrations [65], making it an urgent need to find new classes or combinations of insecticides [35, 17]. In areas where An. funestus is dominant, such as in south-eastern Tanzania, the decisions on which insecticides to be implemented in vector control measures should reflect intensity of resistance in this species, even if it is difficult to find its larvae. Anopheles funestus were resistant up to ten times the WHO-recommended concentration of pyrethroids, clearly indicating that this class of insecticides can no longer be useful in the area and must be urgently replaced by other classes such as organophosphates, against which resistance is not yet detected.
The synergist tests in this study showed complete or almost complete restoration of susceptibility in the malaria vector mosquitoes nearly from all study areas. This full restoration is a likely indicator of metabolic resistance [58, 66] and suggests that ITNs which have both PBO and pyrethroids, such as PermaNet 3.0 [14] and Olyset Plus [13], may be suitable for malaria prevention in these areas, and could potentially provide better protection than standard LLINs [67]. Synergist pre-exposure combined with deltamethrin had a greater restoration in An. funestus than when the synergist was combined with permethrin (Fig. 4), but in both cases there was still substantial restoration. This could be likely due to different resistance levels against the two pyrethroid classes as observed by Rakotoson et al. [68] when An. arabiensis were pre-exposed to PBO. Partial restoration of susceptibility observed in An. funestus mosquitoes might be a sign of multiple metabolic resistance forms or other resistance mechanisms including the target-site mutation [69]. This could also be a manifestation of the demonstrated high intensities of pyrethroid resistance (Fig. 3). These findings are in line with the previous studies on the resistance of malaria vectors to pyrethroids and organochlorides and incomplete susceptibility restoration after the synergist pre-exposure to pyrethroids [68]. Nonetheless, further exploration is needed to identify the specific metabolic enzymes responsible for the observed resistance under biochemical tests. Additionally, the level of these resistant enzymes needs to be assessed using quantitative PCR assays in both An. arabiensis and An. funestus.
Despite largely achieving the stated aims, the findings of this study should be considered only as indicative and not in any way conclusive. This is due to the various methodological limitations faced during the study. First, the overall collection of the specimen was distributed over several months, and may bave been influenced by seasonal variations in resistance, as previously demonstrated [45]. Besides, given the scarcity of An. funestus specimen in some of the villages, the tests used just 120 mosquitoes. Third, this study was the use of wild mosquitoes which may have varying ages, which is an important factor long-demonstrated to impact resistance [7072].
While this way of testing gives a true representation of the natural mosquito population in communities and their ability to withstand insecticidal interventions, it makes it difficult to compare the tests in a conclusive manner. The WHO guidelines recommend the use of age-synchronized F1 generation, 3–5 days old [18]. In this study the challenge was minimized by: (a) collecting the adult female mosquitoes at the edges of the village near potential aquatic habitats, thus maximizing the chances of getting young nulliparous mosquitoes [49], (b) adding an acclimatization period of mosquitoes for 24 h between the actual mosquito collection and the resistance tests, and (c) using the CDC light trap for mosquito collection, thereby capitalizing collection of nulliparous host-seeking mosquitoes [7375]. In addition, the tests did not combine collections from multiple days, but instead used synchronized days for each replicate, thus ensuring that the mosquito ages were approximately similar. It is recognized however that these improvements slightly improved the tests but are not adequate to enable conclusive determination or comparison of resistance levels.
Another limitation was the non-amplification of the samples where 8% (n = 12) of An.arabiensis and 24% (n = 62) of An. funestus complex were unidentifiable. It is possible that either there were polymorphisms in the ITS2 region of rDNA amplified in these assays, which might have been the main contributor of the observed non-amplification (Mapua et al., unpublished data), or there were a few other sibling species for which no primers were available in the assay. Future studies should involve a larger sample size, and possibly individual analysis of specimen to distinguish between species.

Conclusion

Overall, this study has demonstrated that other than the differential importance of malaria vector species and the multiplicity of malaria transmission in different settings, the responsiveness of these vectors towards different insecticides may also vary. In rural south-eastern Tanzania, An. funestus, which now dominates malaria transmission, also indicate stronger resistance to pyrethroids commonly used on ITNs than its counterpart, An. arabiensis. Despite its rarity at aquatic stage, collection methods must endeavour to find this vector with synchronized age and study its resistance profile so that effective interventions can be mounted. Lastly, the study also emphasizes that decisions on which insecticidal interventions to apply should be informed by geographical and species-specific studies rather than generalized studies. In this cases, it appears that PBO-based LLINs and IRS with non pyrethroids, such as organophosphates may be appropriate for now, as the main vectors are still susceptible to these treatments.

Acknowledgements

We thank the research team for their great support and volunteers who were involved in this study. We also thank our project administrator Rukiyah Mohamed Njalambaha for logistical support.
This study was permitted by the Institute Review Board of Ifakara Health Institute IHI/IRB/No: 19–2017 and Medical Research Coordinated Committee of the National Institute for Medical Research of the United Republic of Tanzania NIMR/HQ/R.8c/Vol. I/1185. All study household participants were recruited after signing informed consent forms.
This manuscript has been approved for publication by the Institute for Medical Research of the United Republic of Tanzania NIMR/HQ/P.12 VOL XXX/.

Competing interests

Authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207–11. PubMedPubMedCentralCrossRef Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207–11. PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat WHO. World malaria report 2019. Geneva: World Health Organization; 2019. WHO. World malaria report 2019. Geneva: World Health Organization; 2019.
3.
Zurück zum Zitat Hemingway J, Ranson H, Magill A, Kolaczinski J, Fornadel C, Gimnig J, et al. Averting a malaria disaster: will insecticide resistance derail malaria control? Lancet. 2016;387:1785–8. PubMedPubMedCentralCrossRef Hemingway J, Ranson H, Magill A, Kolaczinski J, Fornadel C, Gimnig J, et al. Averting a malaria disaster: will insecticide resistance derail malaria control? Lancet. 2016;387:1785–8. PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Hemingway J. Resistance: a problem without an easy solution. Pestic Biochem Physiol. 2018;151:73–5. PubMedCrossRef Hemingway J. Resistance: a problem without an easy solution. Pestic Biochem Physiol. 2018;151:73–5. PubMedCrossRef
5.
Zurück zum Zitat The malERA Refresh Consultative Panel on Insecticide and Drug Resistance. malERA: an updated research agenda for insecticide and drug resistance in malaria elimination and eradication. PLoS Med. 2017;14:e1002450. The malERA Refresh Consultative Panel on Insecticide and Drug Resistance. malERA: an updated research agenda for insecticide and drug resistance in malaria elimination and eradication. PLoS Med. 2017;14:e1002450.
6.
Zurück zum Zitat Russell TL, Govella NJ, Azizi S, Drakeley CJ, Kachur SP, Killeen GF. Increased proportions of outdoor feeding among residual malaria vector populations following increased use of insecticide-treated nets in rural Tanzania. Malar J. 2011;10:80. PubMedPubMedCentralCrossRef Russell TL, Govella NJ, Azizi S, Drakeley CJ, Kachur SP, Killeen GF. Increased proportions of outdoor feeding among residual malaria vector populations following increased use of insecticide-treated nets in rural Tanzania. Malar J. 2011;10:80. PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Elliott R. The influence of vector behavior on malaria transmission. Am J Trop Med Hyg. 1972;21:755–63. PubMedCrossRef Elliott R. The influence of vector behavior on malaria transmission. Am J Trop Med Hyg. 1972;21:755–63. PubMedCrossRef
8.
Zurück zum Zitat Sougoufara S, Diédhiou SM, Doucouré S, Diagne N, Sembène PM, Harry M, et al. Biting by Anopheles funestus in broad daylight after use of long-lasting insecticidal nets: a new challenge to malaria elimination. Malar J. 2014;13:125. PubMedPubMedCentralCrossRef Sougoufara S, Diédhiou SM, Doucouré S, Diagne N, Sembène PM, Harry M, et al. Biting by Anopheles funestus in broad daylight after use of long-lasting insecticidal nets: a new challenge to malaria elimination. Malar J. 2014;13:125. PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Monroe A, Moore S, Koenker H, Lynch M, Ricotta E. Measuring and characterizing night time human behaviour as it relates to residual malaria transmission in sub-Saharan Africa: a review of the published literature. Malar J. 2019;18:6. PubMedPubMedCentralCrossRef Monroe A, Moore S, Koenker H, Lynch M, Ricotta E. Measuring and characterizing night time human behaviour as it relates to residual malaria transmission in sub-Saharan Africa: a review of the published literature. Malar J. 2019;18:6. PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Sherrard-Smith E, Skarp JE, Beale AD, Fornadel C, Norris LC, Moore SJ, et al. Mosquito feeding behavior and how it influences residual malaria transmission across Africa. Proc Natl Acad Sci USA. 2019;116:15086–95. PubMedCrossRefPubMedCentral Sherrard-Smith E, Skarp JE, Beale AD, Fornadel C, Norris LC, Moore SJ, et al. Mosquito feeding behavior and how it influences residual malaria transmission across Africa. Proc Natl Acad Sci USA. 2019;116:15086–95. PubMedCrossRefPubMedCentral
12.
Zurück zum Zitat N’Guessan R, Odjo A, Ngufor C, Malone D, Rowland M. A chlorfenapyr mixture net interceptor® G2 shows high efficacy and wash durability against resistant mosquitoes in West Africa. PLoS ONE. 2016;11:e0165925. PubMedPubMedCentralCrossRef N’Guessan R, Odjo A, Ngufor C, Malone D, Rowland M. A chlorfenapyr mixture net interceptor® G2 shows high efficacy and wash durability against resistant mosquitoes in West Africa. PLoS ONE. 2016;11:e0165925. PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Protopopoff N, Mosha JF, Lukole E, Charlwood JD, Wright A, Mwalimu CD, et al. Effectiveness of a long-lasting piperonyl butoxide-treated insecticidal net and indoor residual spray interventions, separately and together, against malaria transmitted by pyrethroid-resistant mosquitoes: a cluster, randomised controlled, two-by-two fact. Lancet. 2018;391:1577–88. PubMedPubMedCentralCrossRef Protopopoff N, Mosha JF, Lukole E, Charlwood JD, Wright A, Mwalimu CD, et al. Effectiveness of a long-lasting piperonyl butoxide-treated insecticidal net and indoor residual spray interventions, separately and together, against malaria transmitted by pyrethroid-resistant mosquitoes: a cluster, randomised controlled, two-by-two fact. Lancet. 2018;391:1577–88. PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Tungu P, Magesa S, Maxwell C, Malima R, Masue D, Sudi W, et al. Evaluation of permanet 3.0 a deltamethrin-PBO combination net against Anopheles gambiae and pyrethroid resistant Culex quinquefasciatus mosquitoes: an experimental hut trial in Tanzania. Malar J. 2010;9:21. PubMedPubMedCentralCrossRef Tungu P, Magesa S, Maxwell C, Malima R, Masue D, Sudi W, et al. Evaluation of permanet 3.0 a deltamethrin-PBO combination net against Anopheles gambiae and pyrethroid resistant Culex quinquefasciatus mosquitoes: an experimental hut trial in Tanzania. Malar J. 2010;9:21. PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Karunaratne P, De Silva P, Weeraratne T, Surendran N. Insecticide resistance in mosquitoes: development, mechanisms and monitoring. Ceylon J Sci. 2018;47:299–309. CrossRef Karunaratne P, De Silva P, Weeraratne T, Surendran N. Insecticide resistance in mosquitoes: development, mechanisms and monitoring. Ceylon J Sci. 2018;47:299–309. CrossRef
16.
Zurück zum Zitat Hemingway J, Ranson H. Insecticide resistance in insect vectors of human disease. Annu Rev Entomol. 2000;45:371–91. PubMedCrossRef Hemingway J, Ranson H. Insecticide resistance in insect vectors of human disease. Annu Rev Entomol. 2000;45:371–91. PubMedCrossRef
17.
Zurück zum Zitat Ranson H. Current and future prospects for preventing malaria transmission via the use of insecticides. Cold Spring Harb Perspect Med. 2017;7:a026823. PubMedPubMedCentralCrossRef Ranson H. Current and future prospects for preventing malaria transmission via the use of insecticides. Cold Spring Harb Perspect Med. 2017;7:a026823. PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat WHO. Test procedures for insecticide resistance monitoring in malaria vector mosquitoes. 2nd ed. Geneva: World Health Organization; 2018a. WHO. Test procedures for insecticide resistance monitoring in malaria vector mosquitoes. 2nd ed. Geneva: World Health Organization; 2018a.
19.
Zurück zum Zitat Brogdon WG, Allister JCMC, Corwin AM, Cordon-rosales C. Independent selection of multiple mechanisms for pyrethroid resistance in Guatemalan Anopheles albimanus (Diptera : Culicidae). J Econ Entomol. 1999;92:298–302. PubMedCrossRef Brogdon WG, Allister JCMC, Corwin AM, Cordon-rosales C. Independent selection of multiple mechanisms for pyrethroid resistance in Guatemalan Anopheles albimanus (Diptera : Culicidae). J Econ Entomol. 1999;92:298–302. PubMedCrossRef
20.
Zurück zum Zitat Vulule JM, Beach RF, Atieli FK. Elevated oxidase and esterase levels associated with permethrin tolerance in Anopheles gambiae from Kenyan villages using permethrin-impregnated nets. Med Vet Entomol. 1999;13:239–44. PubMedCrossRef Vulule JM, Beach RF, Atieli FK. Elevated oxidase and esterase levels associated with permethrin tolerance in Anopheles gambiae from Kenyan villages using permethrin-impregnated nets. Med Vet Entomol. 1999;13:239–44. PubMedCrossRef
21.
Zurück zum Zitat Chouaïbou M, Zivanovic GB, Knox TB, Jamet HP, Bonfoh B. Synergist bioassays: a simple method for initial metabolic resistance investigation of field Anopheles gambiae s.l. populations. Acta Trop. 2014;130:108–11. PubMedPubMedCentralCrossRef Chouaïbou M, Zivanovic GB, Knox TB, Jamet HP, Bonfoh B. Synergist bioassays: a simple method for initial metabolic resistance investigation of field Anopheles gambiae s.l. populations. Acta Trop. 2014;130:108–11. PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Davies TGE, Field LM, Usherwood PNR, Williamson MS. DDT, pyrethrins, pyrethroids and insect sodium channels. IUBMB Life. 2008;59:151–62. CrossRef Davies TGE, Field LM, Usherwood PNR, Williamson MS. DDT, pyrethrins, pyrethroids and insect sodium channels. IUBMB Life. 2008;59:151–62. CrossRef
23.
Zurück zum Zitat Donnelly MJ, Corbel V, Weetman D, Wilding CS, Williamson MS, Black WC IV. Does kdr genotype predict insecticide-resistance phenotype in mosquitoes? Trends Parasitol. 2009;25:213–9. PubMedCrossRef Donnelly MJ, Corbel V, Weetman D, Wilding CS, Williamson MS, Black WC IV. Does kdr genotype predict insecticide-resistance phenotype in mosquitoes? Trends Parasitol. 2009;25:213–9. PubMedCrossRef
24.
Zurück zum Zitat Liu N. Insecticide resistance in mosquitoes: impact, mechanisms, and research directions. Annu Rev Entomol. 2015;60:537–59. PubMedCrossRef Liu N. Insecticide resistance in mosquitoes: impact, mechanisms, and research directions. Annu Rev Entomol. 2015;60:537–59. PubMedCrossRef
25.
Zurück zum Zitat Ingham VA, Anthousi A, Douris V, Harding NJ, Lycett G, Morris M, et al. A sensory appendage protein protects malaria vectors from pyrethroids. Nature. 2020;577:376–80. PubMedCrossRef Ingham VA, Anthousi A, Douris V, Harding NJ, Lycett G, Morris M, et al. A sensory appendage protein protects malaria vectors from pyrethroids. Nature. 2020;577:376–80. PubMedCrossRef
26.
27.
Zurück zum Zitat Cohuet A, Harris C, Robert V, Fontenille D. Evolutionary forces on Anopheles: what makes a malaria vector? Trends Parasitol. 2010;26:130–6. PubMedCrossRef Cohuet A, Harris C, Robert V, Fontenille D. Evolutionary forces on Anopheles: what makes a malaria vector? Trends Parasitol. 2010;26:130–6. PubMedCrossRef
28.
Zurück zum Zitat Kaindoa EW, Matowo NS, Ngowo HS, Mkandawile G, Mmbando A, Finda M, et al. Interventions that effectively target Anopheles funestus mosquitoes could significantly improve control of persistent malaria transmission in south—eastern Tanzania. PLoS ONE. 2017;12:e0177807. PubMedPubMedCentralCrossRef Kaindoa EW, Matowo NS, Ngowo HS, Mkandawile G, Mmbando A, Finda M, et al. Interventions that effectively target Anopheles funestus mosquitoes could significantly improve control of persistent malaria transmission in south—eastern Tanzania. PLoS ONE. 2017;12:e0177807. PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Swai JK, Mmbando AS, Ngowo HS, Odufuwa OG, Finda MF, Mponzi W, et al. Protecting migratory farmers in rural Tanzania using eave ribbons treated with the spatial mosquito repellent, transfluthrin. Malar J. 2019;18:414. PubMedPubMedCentralCrossRef Swai JK, Mmbando AS, Ngowo HS, Odufuwa OG, Finda MF, Mponzi W, et al. Protecting migratory farmers in rural Tanzania using eave ribbons treated with the spatial mosquito repellent, transfluthrin. Malar J. 2019;18:414. PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Finda MF, Limwagu AJ, Ngowo HS, Matowo NS, Swai JK, Kaindoa E, et al. Dramatic decreases of malaria transmission intensities in Ifakara, south-eastern Tanzania since early 2000s. Malar J. 2018;17:362. PubMedPubMedCentralCrossRef Finda MF, Limwagu AJ, Ngowo HS, Matowo NS, Swai JK, Kaindoa E, et al. Dramatic decreases of malaria transmission intensities in Ifakara, south-eastern Tanzania since early 2000s. Malar J. 2018;17:362. PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Takken W, Verhulst NO. Host preferences of blood-feeding mosquitoes. Annu Rev Entomol. 2013;58:433–53. PubMedCrossRef Takken W, Verhulst NO. Host preferences of blood-feeding mosquitoes. Annu Rev Entomol. 2013;58:433–53. PubMedCrossRef
32.
Zurück zum Zitat Kiszewski A, Mellinger A, Spielman A, Malaney P, Sachs SE, Sachs J. A global index representing the stability of malaria transmission. Am J Trop Med Hyg. 2004;70:486–98. PubMedCrossRef Kiszewski A, Mellinger A, Spielman A, Malaney P, Sachs SE, Sachs J. A global index representing the stability of malaria transmission. Am J Trop Med Hyg. 2004;70:486–98. PubMedCrossRef
33.
Zurück zum Zitat Ngowo HS, Kaindoa EW, Matthiopoulos J, Ferguson HM, Okumu FO. Variations in household microclimate affect outdoor-biting behaviour of malaria vectors. Wellcome Open Res. 2017;2:102. PubMedPubMedCentralCrossRef Ngowo HS, Kaindoa EW, Matthiopoulos J, Ferguson HM, Okumu FO. Variations in household microclimate affect outdoor-biting behaviour of malaria vectors. Wellcome Open Res. 2017;2:102. PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Lalji S, Ngondi JM, Thawer NG, Tembo A, Mandike R, Mohamed A, et al. School distribution as keep-up strategy to maintain universal coverage of long-lasting insecticidal nets: implementation and results of a program in southern Tanzania. Glob Health Sci Pract. 2016;4:251–63. PubMedPubMedCentralCrossRef Lalji S, Ngondi JM, Thawer NG, Tembo A, Mandike R, Mohamed A, et al. School distribution as keep-up strategy to maintain universal coverage of long-lasting insecticidal nets: implementation and results of a program in southern Tanzania. Glob Health Sci Pract. 2016;4:251–63. PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Stuck L, Lutambi A, Chacky F, Schaettle P, Kramer K, Mandike R, et al. Can school-based distribution be used to maintain coverage of long-lasting insecticide treated bed nets: evidence from a large scale programme in southern Tanzania? Health Policy Plan. 2017;32:9809. CrossRef Stuck L, Lutambi A, Chacky F, Schaettle P, Kramer K, Mandike R, et al. Can school-based distribution be used to maintain coverage of long-lasting insecticide treated bed nets: evidence from a large scale programme in southern Tanzania? Health Policy Plan. 2017;32:9809. CrossRef
36.
Zurück zum Zitat Bonner K, Mwita A, McElroy PD, Omari S, Mzava A, Lengeler C, et al. Design, implementation and evaluation of a national campaign to distribute nine million free LLINs to children under five years of age in Tanzania. Malar J. 2011;10:73. PubMedPubMedCentralCrossRef Bonner K, Mwita A, McElroy PD, Omari S, Mzava A, Lengeler C, et al. Design, implementation and evaluation of a national campaign to distribute nine million free LLINs to children under five years of age in Tanzania. Malar J. 2011;10:73. PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Renggli S, Mandike R, Kramer K, Patrick F, Brown NJ, McElroy PD, et al. Design, implementation and evaluation of a national campaign to deliver 18 million free long-lasting insecticidal nets to uncovered sleeping spaces in Tanzania. Malar J. 2013;12:85. PubMedPubMedCentralCrossRef Renggli S, Mandike R, Kramer K, Patrick F, Brown NJ, McElroy PD, et al. Design, implementation and evaluation of a national campaign to deliver 18 million free long-lasting insecticidal nets to uncovered sleeping spaces in Tanzania. Malar J. 2013;12:85. PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Gillies MT, Smith A. The effect of a residual house-spraying campaign in east africa on species balance in the Anopheles funestus group the replacement of A. funestus Giles by A rivulorum Leeson. Bull Entomol Res. 1960;51:243–52. CrossRef Gillies MT, Smith A. The effect of a residual house-spraying campaign in east africa on species balance in the Anopheles funestus group the replacement of A. funestus Giles by A rivulorum Leeson. Bull Entomol Res. 1960;51:243–52. CrossRef
39.
Zurück zum Zitat Mabaso MLH, Sharp B, Lengeler C. Historical review of malarial control in southern African with emphasis on the use of indoor residual house-spraying. Trop Med Int Health. 2004;9:846–56. PubMedCrossRef Mabaso MLH, Sharp B, Lengeler C. Historical review of malarial control in southern African with emphasis on the use of indoor residual house-spraying. Trop Med Int Health. 2004;9:846–56. PubMedCrossRef
40.
Zurück zum Zitat Gillies MT, De Meillon B. The Anophelinae of Africa south of the Sahara (Ethiopian Zoogeographical Region). Johannesburg: South African Institute for Medical Research; 1968. Gillies MT, De Meillon B. The Anophelinae of Africa south of the Sahara (Ethiopian Zoogeographical Region). Johannesburg: South African Institute for Medical Research; 1968.
41.
Zurück zum Zitat Lwetoijera DW, Harris C, Kiware SS, Dongus S, Devine GJ, McCall PJ, et al. Increasing role of Anopheles funestus and Anopheles arabiensis in malaria transmission in the Kilombero Valley. Tanzania Malar J. 2014;13:331. PubMedCrossRef Lwetoijera DW, Harris C, Kiware SS, Dongus S, Devine GJ, McCall PJ, et al. Increasing role of Anopheles funestus and Anopheles arabiensis in malaria transmission in the Kilombero Valley. Tanzania Malar J. 2014;13:331. PubMedCrossRef
42.
Zurück zum Zitat Russell TL, Lwetoijera DW, Maliti D, Chipwaza B, Kihonda J, Charlwood JD, et al. Impact of promoting longer-lasting insecticide treatment of bed nets upon malaria transmission in a rural Tanzanian setting with pre-existing high coverage of untreated nets. Malar J. 2010;9:187. PubMedPubMedCentralCrossRef Russell TL, Lwetoijera DW, Maliti D, Chipwaza B, Kihonda J, Charlwood JD, et al. Impact of promoting longer-lasting insecticide treatment of bed nets upon malaria transmission in a rural Tanzanian setting with pre-existing high coverage of untreated nets. Malar J. 2010;9:187. PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Bayoh MN, Mathias DK, Odiere MR, Mutuku FM, Kamau L, Gimnig JE, et al. Anopheles gambiae: Historical population decline associated with regional distribution of insecticide-treated bed nets in western Nyanza Province. Kenya Malar J. 2010;9:62. PubMedCrossRef Bayoh MN, Mathias DK, Odiere MR, Mutuku FM, Kamau L, Gimnig JE, et al. Anopheles gambiae: Historical population decline associated with regional distribution of insecticide-treated bed nets in western Nyanza Province. Kenya Malar J. 2010;9:62. PubMedCrossRef
44.
Zurück zum Zitat WHO. Global report on insecticide resistance in malaria vectors: 2010–2016 Global Malaria Programme. Geneva: World Health Organization; 2018b. WHO. Global report on insecticide resistance in malaria vectors: 2010–2016 Global Malaria Programme. Geneva: World Health Organization; 2018b.
45.
Zurück zum Zitat Matowo NS, Abbasi S, Munhenga G, Tanner M, Mapua SA, Oullo D, et al. Fine-scale spatial and temporal variations in insecticide resistance in Culex pipiens complex mosquitoes in rural south-eastern Tanzania. Parasit Vectors. 2019;12:413. PubMedPubMedCentralCrossRef Matowo NS, Abbasi S, Munhenga G, Tanner M, Mapua SA, Oullo D, et al. Fine-scale spatial and temporal variations in insecticide resistance in Culex pipiens complex mosquitoes in rural south-eastern Tanzania. Parasit Vectors. 2019;12:413. PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Sriwichai P, Karl S, Samung Y, Sumruayphol S, Kiattibutr K, Payakkapol A, et al. Evaluation of CDC light traps for mosquito surveillance in a malaria endemic area on the Thai-Myanmar border. Parasit Vectors. 2015;8:636. PubMedPubMedCentralCrossRef Sriwichai P, Karl S, Samung Y, Sumruayphol S, Kiattibutr K, Payakkapol A, et al. Evaluation of CDC light traps for mosquito surveillance in a malaria endemic area on the Thai-Myanmar border. Parasit Vectors. 2015;8:636. PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Mboera LEG, Kihonda J, Braks MAH, Knols BGJ. Influence of centers for disease control light trap position, relative to a human-baited bed net, on catches of Anopheles gambiae and Culex quinquefasciatus in Tanzania. Am J Trop Med Hyg. 1998;59:595–6. PubMedCrossRef Mboera LEG, Kihonda J, Braks MAH, Knols BGJ. Influence of centers for disease control light trap position, relative to a human-baited bed net, on catches of Anopheles gambiae and Culex quinquefasciatus in Tanzania. Am J Trop Med Hyg. 1998;59:595–6. PubMedCrossRef
51.
Zurück zum Zitat Gillies MT, Coetzee M. A Supplement to the Anophelinae of the South of the Sahara (Afrotropical Region). Publ South African Inst Med Res. 1987;55:1–143. Gillies MT, Coetzee M. A Supplement to the Anophelinae of the South of the Sahara (Afrotropical Region). Publ South African Inst Med Res. 1987;55:1–143.
52.
Zurück zum Zitat Koekemoer LL, Kamau L, Hunt RH, Coetzee M. A cocktail polymerase chain reaction assay to identify members of the Anopheles funestus (Diptera : Culicida) group. Am J Trop Med Hyg. 2002;66:804–11. PubMedCrossRef Koekemoer LL, Kamau L, Hunt RH, Coetzee M. A cocktail polymerase chain reaction assay to identify members of the Anopheles funestus (Diptera : Culicida) group. Am J Trop Med Hyg. 2002;66:804–11. PubMedCrossRef
53.
Zurück zum Zitat Collins FH. Identification of single specimen of the Anopheles gambiae complex by the polymerase chain reaction. Am J Trop Med Hyg. 1993;49:520–9. PubMedCrossRef Collins FH. Identification of single specimen of the Anopheles gambiae complex by the polymerase chain reaction. Am J Trop Med Hyg. 1993;49:520–9. PubMedCrossRef
54.
Zurück zum Zitat Constans A. Real-time gel documentation: the KODAK Gel Logic 100 Imaging System offers gel imaging and analysis for high-throughput labs (Lab Consumer). Scientist. 2002;16:52. Constans A. Real-time gel documentation: the KODAK Gel Logic 100 Imaging System offers gel imaging and analysis for high-throughput labs (Lab Consumer). Scientist. 2002;16:52.
55.
Zurück zum Zitat Abbott WS. A method of computing the effectiveness of an insecticide. J Econ Entomol. 1925;18:265–7. CrossRef Abbott WS. A method of computing the effectiveness of an insecticide. J Econ Entomol. 1925;18:265–7. CrossRef
57.
Zurück zum Zitat Martinez-Torres D, Chandre F, Williamson MS, Darriet F, Bergé JB, Devonshire AL, et al. Molecular characterization of pyrethroid knockdown resistance (kdr) in the major malaria vector Anopheles gambiae s.s. Insect Mol Biol. 1998;7:179–84. PubMedCrossRef Martinez-Torres D, Chandre F, Williamson MS, Darriet F, Bergé JB, Devonshire AL, et al. Molecular characterization of pyrethroid knockdown resistance (kdr) in the major malaria vector Anopheles gambiae s.s. Insect Mol Biol. 1998;7:179–84. PubMedCrossRef
58.
Zurück zum Zitat Matowo NS, Munhenga G, Tanner M, Coetzee M, Feringa WF, Ngowo HS, et al. Fine-scale spatial and temporal heterogeneities in insecticide resistance profiles of the malaria vector, Anopheles arabiensis in rural south-eastern Tanzania. Wellcome Open Res. 2017;2:96. PubMedPubMedCentralCrossRef Matowo NS, Munhenga G, Tanner M, Coetzee M, Feringa WF, Ngowo HS, et al. Fine-scale spatial and temporal heterogeneities in insecticide resistance profiles of the malaria vector, Anopheles arabiensis in rural south-eastern Tanzania. Wellcome Open Res. 2017;2:96. PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Yadouleton AWM, Asidi A, Djouaka RF, Brama J, Agossou CD, Akogbeto MC. Development of vegetable farming: a cause of the emergence of insecticide resistance in populations of Anopheles gambiae in urban areas of Benin. Malar J. 2009;8:103. PubMedPubMedCentralCrossRef Yadouleton AWM, Asidi A, Djouaka RF, Brama J, Agossou CD, Akogbeto MC. Development of vegetable farming: a cause of the emergence of insecticide resistance in populations of Anopheles gambiae in urban areas of Benin. Malar J. 2009;8:103. PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Menze BD, Riveron JM, Ibrahim SS, Irving H, Antonio-Nkondjio C, Awono-Ambene PH, et al. Multiple insecticide resistance in the malaria vector Anopheles funestus from Northern Cameroon is mediated by metabolic resistance alongside potential target site insensitivity mutations. PLoS ONE. 2016;11:e0163261. PubMedPubMedCentralCrossRef Menze BD, Riveron JM, Ibrahim SS, Irving H, Antonio-Nkondjio C, Awono-Ambene PH, et al. Multiple insecticide resistance in the malaria vector Anopheles funestus from Northern Cameroon is mediated by metabolic resistance alongside potential target site insensitivity mutations. PLoS ONE. 2016;11:e0163261. PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Djouaka R, Riveron JM, Yessoufou A, Tchigossou G, Akoton R, Irving H, et al. Multiple insecticide resistance in an infected population of the malaria vector Anopheles funestus in Benin. Parasit Vectors. 2016;9:453. PubMedPubMedCentralCrossRef Djouaka R, Riveron JM, Yessoufou A, Tchigossou G, Akoton R, Irving H, et al. Multiple insecticide resistance in an infected population of the malaria vector Anopheles funestus in Benin. Parasit Vectors. 2016;9:453. PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Djouaka RJ, Atoyebi SM, Tchigossou GM, Riveron JM, Irving H, Akoton R, et al. Evidence of a multiple insecticide resistance in the malaria vector Anopheles funestus in South West Nigeria. Malar J. 2016;15:565. PubMedPubMedCentralCrossRef Djouaka RJ, Atoyebi SM, Tchigossou GM, Riveron JM, Irving H, Akoton R, et al. Evidence of a multiple insecticide resistance in the malaria vector Anopheles funestus in South West Nigeria. Malar J. 2016;15:565. PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Mzilahowa T, Chiumia M, Mbewe RB, Uzalili VT, Banda ML, Kutengule A, et al. Increasing insecticide resistance in Anopheles funestus and Anopheles arabiensis in Malawi, 2011–2015. Malar J. 2016;15:563. PubMedPubMedCentralCrossRef Mzilahowa T, Chiumia M, Mbewe RB, Uzalili VT, Banda ML, Kutengule A, et al. Increasing insecticide resistance in Anopheles funestus and Anopheles arabiensis in Malawi, 2011–2015. Malar J. 2016;15:563. PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Riveron JM, Osae M, Egyir-Yawson A, Irving H, Ibrahim SS, Wondji CS. Multiple insecticide resistance in the major malaria vector Anopheles funestus in southern Ghana: implications for malaria control. Parasit Vectors. 2016;9:504. PubMedPubMedCentralCrossRef Riveron JM, Osae M, Egyir-Yawson A, Irving H, Ibrahim SS, Wondji CS. Multiple insecticide resistance in the major malaria vector Anopheles funestus in southern Ghana: implications for malaria control. Parasit Vectors. 2016;9:504. PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Toé KH, Jones CM, N’fale S, Ismai HM, Dabiré RK, Ranson H. Increased pyrethroid resistance in malaria vectors and decreased bed net effectiveness Burkina Faso. Emerg Infect Dis. 2014;20:1691–6. PubMedPubMedCentralCrossRef Toé KH, Jones CM, N’fale S, Ismai HM, Dabiré RK, Ranson H. Increased pyrethroid resistance in malaria vectors and decreased bed net effectiveness Burkina Faso. Emerg Infect Dis. 2014;20:1691–6. PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Zoh DD, Ahoua Alou LP, Toure M, Pennetier C, Camara S, Traore DF, et al. The current insecticide resistance status of Anopheles gambiae (sl) (Culicidae) in rural and urban areas of Bouaké. Côte d’Ivoire Parasit Vectors. 2018;11:118. PubMedCrossRef Zoh DD, Ahoua Alou LP, Toure M, Pennetier C, Camara S, Traore DF, et al. The current insecticide resistance status of Anopheles gambiae (sl) (Culicidae) in rural and urban areas of Bouaké. Côte d’Ivoire Parasit Vectors. 2018;11:118. PubMedCrossRef
67.
Zurück zum Zitat Gleave K, Lissenden N, Richardson M, Choi L, Ranson H. Piperonyl butoxide (PBO) combined with pyrethroids in insecticide-treated nets to prevent malaria in Africa. Cochrane Database Syst Rev. 2018;1101:2776. Gleave K, Lissenden N, Richardson M, Choi L, Ranson H. Piperonyl butoxide (PBO) combined with pyrethroids in insecticide-treated nets to prevent malaria in Africa. Cochrane Database Syst Rev. 2018;1101:2776.
68.
Zurück zum Zitat Rakotoson JD, Fornadel CM, Belemvire A, Norris LC, George K, Caranci A, et al. Insecticide resistance status of three malaria vectors, Anopheles gambiae (s.l.), An. funestus and An. mascarensis, from the south, central and east coasts of Madagascar. Parasit Vectors. 2017;10:396. PubMedPubMedCentralCrossRef Rakotoson JD, Fornadel CM, Belemvire A, Norris LC, George K, Caranci A, et al. Insecticide resistance status of three malaria vectors, Anopheles gambiae (s.l.), An. funestus and An. mascarensis, from the south, central and east coasts of Madagascar. Parasit Vectors. 2017;10:396. PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Nwane P, Etang J, Chouabou M, Toto JC, Koffi A, Mimpfoundi R, et al. Multiple insecticide resistance mechanisms in Anopheles gambiae s.l. populations from Cameroon Central Africa. Parasit Vectors. 2013;6:41. PubMedPubMedCentralCrossRef Nwane P, Etang J, Chouabou M, Toto JC, Koffi A, Mimpfoundi R, et al. Multiple insecticide resistance mechanisms in Anopheles gambiae s.l. populations from Cameroon Central Africa. Parasit Vectors. 2013;6:41. PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Kulma K, Saddler A, Koella JC. Effects of age and larval nutrition on phenotypic expression of insecticide-resistance in Anopheles mosquitoes. PLoS ONE. 2013;8:e58322. PubMedPubMedCentralCrossRef Kulma K, Saddler A, Koella JC. Effects of age and larval nutrition on phenotypic expression of insecticide-resistance in Anopheles mosquitoes. PLoS ONE. 2013;8:e58322. PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Chouaibou MS, Chabi J, Bingham GV, Knox TB, N’Dri L, Kesse NB, et al. Increase in susceptibility to insecticides with aging of wild Anopheles gambiae mosquitoes from Côte d’Ivoire. BMC Infect Dis. 2012;12:214. PubMedPubMedCentralCrossRef Chouaibou MS, Chabi J, Bingham GV, Knox TB, N’Dri L, Kesse NB, et al. Increase in susceptibility to insecticides with aging of wild Anopheles gambiae mosquitoes from Côte d’Ivoire. BMC Infect Dis. 2012;12:214. PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Glunt KD, Thomas MB, Read AF. The effects of age, exposure history and malaria infection on the susceptibility of Anopheles mosquitoes to low concentrations of pyrethroid. PLoS ONE. 2011;6:e24968. PubMedPubMedCentralCrossRef Glunt KD, Thomas MB, Read AF. The effects of age, exposure history and malaria infection on the susceptibility of Anopheles mosquitoes to low concentrations of pyrethroid. PLoS ONE. 2011;6:e24968. PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Mboera LE. Sampling techniques for adult Afrotropical malaria vectors and their reliability in the estimation of entomological inoculation rate. Tanzan Health Res Bull. 2005;7:117–24. PubMed Mboera LE. Sampling techniques for adult Afrotropical malaria vectors and their reliability in the estimation of entomological inoculation rate. Tanzan Health Res Bull. 2005;7:117–24. PubMed
74.
Zurück zum Zitat Sadanandane C, Jambulingam P, Subramanian S. Role of modified CDC miniature light-traps as an alternative method for sampling adult anophelines (Diptera: Culicidae) in the National Mosquito Surveillance Programme in India. Bull Entomol Res. 2004;94:55–63. PubMedCrossRef Sadanandane C, Jambulingam P, Subramanian S. Role of modified CDC miniature light-traps as an alternative method for sampling adult anophelines (Diptera: Culicidae) in the National Mosquito Surveillance Programme in India. Bull Entomol Res. 2004;94:55–63. PubMedCrossRef
75.
Zurück zum Zitat Lines JD, Curtis CF, Wilkes TJ, Njunwa KJ. Monitoring human-biting mosquitoes (Diptera: Culicidae) in Tanzania with light-traps hung beside mosquito nets. Bull Entomol Res. 1991;81:77–84. CrossRef Lines JD, Curtis CF, Wilkes TJ, Njunwa KJ. Monitoring human-biting mosquitoes (Diptera: Culicidae) in Tanzania with light-traps hung beside mosquito nets. Bull Entomol Res. 1991;81:77–84. CrossRef
Metadaten
Titel
Comparative assessment of insecticide resistance phenotypes in two major malaria vectors, Anopheles funestus and Anopheles arabiensis in south-eastern Tanzania
verfasst von
Polius G. Pinda
Claudia Eichenberger
Halfan S. Ngowo
Dickson S. Msaky
Said Abbasi
Japhet Kihonda
Hamis Bwanaly
Fredros O. Okumu
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2020
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-020-03483-3

Weitere Artikel der Ausgabe 1/2020

Malaria Journal 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.