Blog

  • The Toxicity of Dimethyl Sulfoxide Combined with Alkylating Agent MNU

    The Toxicity of Dimethyl Sulfoxide Combined with Alkylating Agent MNU

    Introduction

    Dimethyl sulfoxide (DMSO) is a highly polar, organic compound that is widely utilized in scientific and medical researches.1,2 Known for its unique ability to penetrate biological membranes, DMSO is often used as a solvent to facilitate the absorption of a wide variety of chemical compounds, including drugs, toxins, and other substances. This characteristic makes DMSO an invaluable tool in laboratory experiments, particularly in drug delivery systems and pharmacological studies.3 In particular, DMSO’s ability to increase the permeability of biological membranes can influence the pharmacokinetics and biodistribution of co-administered substances, potentially altering their efficacy and safety profiles. This is especially relevant in toxicological studies, where DMSO is frequently used as a vehicle solvent to solubilize poorly water-soluble compounds. However, its capacity to enhance the absorption of compounds into biological tissues also raises concerns about its potential to exacerbate the toxic effects of those compounds, as high concentrations or prolonged exposure may lead to adverse effects such as hemolysis, allergic reactions, and genotoxicity.

    One of the critical areas where DMSO’s solvent properties are frequently leveraged is in the use of chemical agents such as alkylating agents, which have been extensively studied for their ability to modify the structure and function of biological molecules, particularly DNA. Alkylating agents, including N-methyl-N-nitrosourea (MNU), one of the most commonly used alkylating agents, has been widely utilized in cancer research and retinitis pigmentosa modeling.4,5 These agents could covalently bind to DNA, leading to significant alterations in its structure and function. Specifically, they exert their effects by transferring alkyl groups to DNA, which can cause mutations, chromosomal damage, and eventually carcinogenesis or apoptosis of retinal photoreceptors. MNU, in particular, is a powerful mutagen and carcinogen that has been shown to induce a variety of cancers in laboratory animals, including lymphomas, leukemias, and solid tumors in various organs. Its high reactivity and ability to penetrate biological membranes make it an effective agent for inducing genetic damage in a wide range of cell types. Besides, the effectiveness of MNU as a research tool for studying retinitis pigmentosa has made it a cornerstone in many ocular experimental protocols.6,7 In details, by selectively targeting retinal photoreceptors, MNU allows researchers to mimic the progressive degeneration seen in human retinitis pigmentosa, providing a valuable model for testing potential therapeutic interventions.

    While DMSO is often regarded as relatively safe when used at low concentrations, its interaction with other chemicals and the subsequent effects on the health of experimental animals require careful consideration. Low levels of DMSO may interact with cellular processes or metabolic pathways in ways that are not yet fully understood. Studies involving DMSO as a vehicle solvent for chemical agents need to take into account the potential for DMSO to alter the pharmacokinetics and toxicity profiles of the compounds being studied. It may influence absorption rates, distribution within tissues, and elimination half-lives, potentially leading to unexpected outcomes. For example, DMSO was shown to increase the permeability of the blood-brain barrier and enhance the systemic distribution,8 which could result in higher concentrations reaching sensitive areas such as the central nervous system, thereby amplifying their therapeutic or toxic effects. However, despite the well-documented uses of DMSO and MNU in research,9 the combined effects of these substances on biological systems have not been fully elucidated, particularly in terms of long-term consequences or subtle interactions that may only become apparent after prolonged exposure. The potential of DMSO to enhance the absorption of MNU into biological tissues might lead to a more pronounced toxicological effect than when either agent is administered alone. This could result in increased DNA damage or oxidative stress, which are critical factors in carcinogenesis and tissue injury. Given that alkylating agents like MNU can induce significant DNA damage, it is important to assess the overall toxicity and safety of these compounds of DSMO and MNU, particularly in animal studies, to ensure that experimental protocols adequately account for potential risks and to establish safe dosage ranges for future research applications.

    The main goal of this study was to investigate the combined toxicity of DMSO and MNU in a controlled animal model. Specifically, this research aimed to assess the impact of DMSO on the toxicity and mortality associated with MNU exposure in C57BL/6J mice, which are commonly used in ocular researches. By varying the doses of MNU and administering it in combination with DMSO, this study provided preliminary data on the synergistic effects of these two compounds. Additionally, the study assessed whether DMSO, by facilitating the absorption of MNU, exacerbated the toxicological outcomes, including changes in activity levels, health status, and overall survival rates in the mice.

    Methods

    Animal Models

    A total of 12 male SPF C57BL/6J mice, aged 8–10 weeks, with an averaged body weight range of 20–25 grams, were selected for this experiment. These mice were obtained from the Laboratory Animal Center of Fuzong Clinical Medical College of Fujian Medical University (License number.: SYXK2018006) and were allowed to acclimate to the laboratory environment for a period of 3 days before the commencement of the study. The mice were housed in standard laboratory conditions, with a controlled room temperature of 22°C ± 2°C and a relative humidity of 50–60%. The animals were maintained on a 12-hour light/dark cycle, with lights on at 7:00 AM and off at 7:00 PM. The mice were provided with standard rodent chow and clean, filtered water ad libitum throughout the experiment. The cages were kept in an environmentally controlled room to minimize external stressors, and the mice were allowed unrestricted access to their environment in order to reduce potential confounding factors.

    After a period of 3 days of accommodation, the mice were randomly assigned to one of three experimental groups, with four mice per group. The groups were: the DMSO control group (D), the low-dose MNU group (L), and the high-dose MNU group (H). We confirmed that ethical and legal approval was obtained from the Experimental Animal Care and Use Committee of Fuzong Clinical Medical College of Fujian Medical University (Ethics committee number: 201923), prior to the commencement of the study. Besides, all animal experiments and procedures were performed in accordance with the ethical guidelines and regulations established by Fuzong Clinical Medical College of Fujian Medical University and our nation. The overall design of our study was showed in the Graphic Abstract of the Study.

    Group Design and Treatment Protocol

    1. DMSO Control Group (D): The D group, serving as the vehicle control, received intraperitoneal (IP) injections of pure DMSO (100% concentration). Each mouse was administered 12 mL of DMSO per kilogram of body weight. This control treatment was designed to assess the baseline effects of DMSO, ensuring that any observed effects in the other groups could be attributed specifically to the MNU treatment, rather than the vehicle itself.
    2. Low-dose MNU Group (L): The L group received a DMSO solution containing 40 mg/kg of MNU,10 the potent genotoxic agent. MNU was dissolved in DMSO to obtain the desired concentration, and the injection volume for all mice was maintained at 12 mL/kg body weight. In greater details, we firstly calculated the required amount of MNU. For example, for a dose of 40 mg/kg in 3 mice weighing 30 g each, the amount of MNU needed would be MNU = 40×10 × 0.03 = 12 mg. Then, we calculated the corresponding concentration of the MNU solution for injection at a volume of 12 mL/kg. If the MNU was administered at 40 mg/kg and injected at a volume of 12 mL/kg, the corresponding concentration of the MNU solution should be 10/3 mg/mL. Thus, 12 mg of MNU would require a volume of DMSO of 3.6 mL. The solution was then stored at 4°C for later use, and prepared immediately before use. The purpose of this group was to examine the effects of a low-dose MNU exposure on the mice, focusing on toxicological effects and any changes in behavior or general health that could result from this exposure level.
    3. High-dose MNU Group (H): The H group received a higher dose of MNU, specifically 60 mg/kg,10 in the DMSO solution. Similar to the L group, the MNU solution was prepared by dissolving MNU in DMSO, and the injection volume was 12 mL/kg of body weight. This higher dose aimed to assess the potential acute toxicity and mortality associated with a more severe exposure to MNU. This group was expected to show more pronounced toxicological effects, including possible lethality, and served as an extreme comparison for the lower dose.

    Injection Procedure

    All injections were administered via intraperitoneal injection, a standard route for delivering substances that need to be absorbed into the body quickly. The injections were carefully carried out by an experienced personnel to ensure precise and consistent dosages across all animals. The animals were weighed immediately prior to injection to ensure accurate dosing, and each mouse was handled gently to reduce stress and minimize discomfort.

    Monitoring and Observations

    Following treatment, the mice were closely monitored for any immediate or delayed signs of distress, toxicity, or adverse effects. The following parameters were tracked throughout the study:

    1. Body Weight: Mice were weighed several timepoints (Timepoint 1 (T1): at the time of purchase (ie, before the 3-day pre-adaptation feeding); T2: before injection (ie, after the 3-day pre-adaptation feeding); T3: at 24 hours post-injection; T4: at 48 hours post-injection) to monitor any significant changes in body weight, which could indicate adverse effects such as dehydration, malnutrition, or systemic toxicity. A marked decrease in body weight was considered a key indicator of toxicity,11,12 and the rate of weight loss was recorded for comparison across groups.
    2. Activity Levels and scoring: Activity levels were observed both during the light and dark cycles at several timepoints (T1: before injection; T2: at 2 hours post-injection; T3: at 24 hours post-injection; T4: at 36 hours post-injection; T5: at 48 hours post-injection). General locomotor activity was evaluated through simple observational methods, such as monitoring the frequency of walking, movement across the cage, and exploratory behavior. Mice were also monitored for signs of lethargy or reduced activity, which are common indicators of stress or illness.13 Additionally, behaviors like body stretching and body posture were noted, as these could reflect changes in the mice’s physical condition or discomfort.14,15 Besides, fur condition was examined daily to assess any signs of stress or health decline. Deterioration in fur quality, such as loss of fur, roughness (or called the ruffled fur), or lack of grooming behavior, was considered an important indicator of poor health or systemic effects of the treatment.15,16 In our study, the activity score was primarily based on three key domains: the mice’s activity alertness (or the locomotor activity), body extension or posture, and the condition of the fur. A total of 9 points were assigned, with 3 points allocated to each domain. The corresponding scores were assessed and recorded by one certain person, and then were stored for later statistical analysis.
    3. Mortality and Survival curve: Mortality was monitored at several timepoints (T1: before injection; T2: at 2 hours post-injection; T3: at 24 hours post-injection; T4: at 36 hours post-injection; T5: at 48 hours post-injection). Any deaths within this period were carefully observed and the number of mice in each group were recorded. Furthermore, the surviving time of mice in each group was recorded and the survival curves were created based on the number of surviving mice and their survival time.17 Further Kaplan-Meier survival curve analysis was then performed.18

    Statistical Analysis

    Due to the small sample size of 4 animals (less than 5 animals) per group, the non-parametric statistical method, the Bootstrap test, was employed to compare the groups.19 This special test was chosen as it does not rely on distributional assumptions, making it more appropriate for small sample sizes.20 The data were presented as mean ± standard deviation. In the Bootstrap method, the calculation of the P-value is based on the distribution of resampled data and does not directly provide a standard P-value. Although there is no direct P-value, the significance can be indirectly assessed through the following approach by examining the confidence interval. If the confidence interval of the Bootstrap results does not include zero, this typically indicates that the difference is significant. The survival analysis was conducted using the Kaplan-Meier survival curves. The statistical analysis was performed using SPSS 27.0 software, and survival curves were generated and analyzed using Log-rank tests. Figures were generated using GraphPad 5.01 software.

    Results

    Pre-Injection Status and Parameters

    Before the injection, the mice in all experimental groups were closely monitored. The 95% confidence interval of difference in the body weight of mice revealed by the Bootstrap analysis between the D and L, D and H, L and H groups, were (−3.16, 2.66), (−3.36, 2.36) and (−2.25, 1.75) respectively. No significant statistical differences were found in terms of body weight, as the confidence interval of the Bootstrap results included zero (Figure 1). The 95% confidence interval of difference in the volume of DMSO injected between the D and L, D and H, L and H groups, were (−0.019,0.012), (−0.025, 0.012) and (−0.016, 0.007) respectively. No significant statistical differences were found as the confidence interval of the Bootstrap results included zero (Figure 2). It suggested that the experimental groups were well-matched at baseline and there was no noticeable variation in these parameters before treatment.

    Figure 1 Body Weight of Each Group of Mice Before Injection. Bar graph showed the body weight of mice in the DMSO, low-dose MNU, and high-dose MNU groups before the injection. There were no significant differences in body weight between the groups (The confidence interval of the Bootstrap results included zero). D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group. N.S.: no statistical difference.

    Figure 2 Volume of DMSO Injected in Each Group of Mice. Bar graph illustrated the volume of DMSO administered to each group (DMSO, low-dose MNU, high-dose MNU). The injection volume was consistent across all groups, at 12 mL/kg body weight, with no significant differences (The confidence interval of the Bootstrap results included zero). D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group. N.S.: no statistical difference.

    Regarding the MNU injection, the dose administered to the mice in the H group was significantly higher than that given to L group (the 95% confidence interval of difference was 0.076–0.096), which was in turn higher than the dose given to the D group (the 95% confidence interval of difference was 0.160–0.166). The statistical analysis confirmed that the differences in MNU doses among the groups were highly significant, as the confidence interval of the Bootstrap results did not include zero (Figure 3). Prior to the injection, all mice in the study exhibited strong mobility, alertness, and a relaxed posture, with their fur appearing glossy and well-groomed, indicating overall good health.

    Figure 3 MNU Dose Injected in Each Group of Mice. Bar graph showed the dose of MNU injected in the DMSO (D), low-dose (L) and high-dose (H) MNU groups. The high-dose group (60 mg/kg) received a significantly higher MNU dose compared to the low-dose group (40 mg/kg) and the DMSO group (0 mg/kg) (The confidence interval of the Bootstrap results did not include zero). D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group. None: not any quality of MNU; **: a significant difference existed.

    Survival Counts and Survival Curves Analysis

    Following the injection, the survival rates varied across the different groups. Two hours after the injection, all mice in all the experimental groups, except for one mouse in the D group, displayed reduced mobility, decreased alertness, and a hunched posture. After 12 hours, mortality of the mice in all groups began to increase, with one mouse in the D group and two mice in the L group being dead. By 24 hours, the mortality rate continued to rise, as another one mouse in the D group, and two mice in both the L and H groups died. After 36 hours, another one mouse in the D group had died, while one mouse in the D group had regained increased activity. The other mice exhibited the poor mobility and the hunched posture. At this point, all mice in the L and H groups had died, leaving only two surviving individuals in D group. After 48 hours, the recovering mouse in the D group showed significant differences in the survival, which remained alive, exhibiting normal walking ability and alertness. Meanwhile the other mice in the D group had died out (Figure 4).

    Figure 4 Number of Each Group of Mice at Different Time Points and the Survival Curves. (A) Number of each group of mice at different time points. (B) The survival curves of each group of mice at different time points. The number of mice in each group with the survival curve displayed the number of surviving mice at various time points after injection in the DMSO, low-dose MNU, and high-dose MNU groups. Mortality increased over time, with the low-dose and high-dose groups experiencing complete mortality by 36 hours. The DMSO group displayed a smaller mortality, with one mouse surviving at 48 hours. T1: before injection; T2: at 12 hours post-injection; T3: at 24 hours post-injection; T4: at 36 hours post-injection; T5: at 48 hours post-injection. D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group.

    Body Weight Changes

    Throughout the course of the study, no significant differences in the changes of body weight were found in the surviving mice across all groups in all the timepoints of observation. Despite the varying mortality rates and changes in activity levels among all groups, the body weight of the surviving mice remained stable in the D, L and H groups. This might suggest that the physiological effects of the treatment did not result in substantial weight loss among the survivors during the observation period (Figure 5).

    Figure 5 Changes in Body Weight of Each Group of Mice After Injection. Line graph showed the changes in body weight over time in the DMSO, low-dose MNU, and high-dose MNU groups after injection. No significant differences in body weight changes were observed among the groups during the observation period. Timepoint 1 (T1): at the time of purchase (ie, before the 3-day pre-adaptation feeding).; T2: before injection (i.e., after the 3-day pre-adaptation feeding); T3: at 24 hours post-injection; T4: at 48 hours post-injection. D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group.

    Activity Levels Changes

    In terms of activity, two hours after the injection, all mice in the experimental groups showed a marked decline in activity levels. They appeared less alert, displayed a hunched posture, and had reduced physical activity compared to their pre-injection status. As the hours passed, the decline in activity became more pronounced. By 24 hours, some mice in all the groups exhibited trembling, and their activity levels were further reduced. At 36 hours, one mouse in the D group managed to regain increased activity, while others showed signs of poor mobility and distress. No significant differences in activity levels scores were found in the surviving mice across all groups. By the 48-hour mark, the only surviving mouse in the D group displayed the normal walking ability and alertness, while all the other mice in the D, L, and H groups had died out (Figures 6 and 7).

    Figure 6 Changes in Activity Scores of Each Group of Mice After Injection. Line graph illustrated the changes in activity scores (measuring mobility, posture, and alertness) over time in each group after injection. Significant reductions in activity were observed in the low-dose and high-dose MNU groups, with severe declines noted by 24 hours. The DMSO group showed some recovery by 36 hours, though further decline was observed at 48 hours. T1: before injection; T2: at 2 hours post-injection; T3: at 24 hours post-injection; T4: at 36 hours post-injection; T5: at 48 hours post-injection. D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group.

    Figure 7 Representative Images of the Activity Levels and Mental Status of Mice at Different Time Points in Each Group. Red arrows indicated mice with reduced activity and a hunched posture, while blue arrows highlighted mice with a good mental status, increased activity, high alertness, and the ability to move freely. Black arrows indicated the dead mice. T1: at 2 hours post-injection; T2: at 24 hours post-injection; T3: at 36 hours post-injection; T4: at 48 hours post-injection. D: the DMSO control group; L: the low-dose MNU group; H: the high-dose MNU group.

    Discussion

    In our study, male C57BL/6J mice were divided into three groups, the D, L and H groups. Mice were monitored for changes in body weight, activity levels, and mortality. Our results showed no significant differences in body weight among the groups prior to injection. Mortality rate was relative increased with injection of DMSO and MNU combination, while the DMSO alone could also led to a high mortality. All mice in the L and H groups had died out by 36 hours, while only one mouse in the D group survived, regaining a normal activity by 48 hours. All mice exhibited a reduced mobility and a hunched posture, with the activity declining progressively, after injection of either the DMSO or the DMSO-MNU combination.

    In greater details, the mice with only the DMSO injection showed a noticeable decline in activity, reduced alertness, and signs of physical distress such as, a hunched posture, the limb curling and dull fur within two hours after injection. These symptoms indicated the presence of toxic effects. Subsequently, the animal deaths were observed, with only one mouse managed to recover and regain normal vitality at the end. The above results suggested that DMSO had a clear toxic effect on animals, with the potential for lethality or mortality. The toxicity of DMSO observed in this study were consistent with findings reported in other researches.21 DMSO was shown to provoke toxic reactions in the body, which might include a variety of adverse effects.22 Several studies also highlighted the toxicological risks associated with DMSO, indicating that it could significantly alter physiological processes and lead to behavioral changes.23 In particular, the impact of 0.1–10% DMSO concentrations on behavior of aquatic model species has also been noted.24 The DMSO concentrations under 1% did not cause statistically significant mortality, but did induce clear signs of stress, reduced locomotion, and impaired responses to stimuli. Given all these observed effects, it is essential for researchers to be cautious when using DMSO in experimental settings, ensuring that proper controls and safety protocols are in place to safeguard the well-being of animal subjects. It is critical that appropriate dilution of DMSO concentrations is conducted in related studies to minimize potential harm. Interestingly, the observation that one mouse with only the DMSO injection showed activity recovery at 36 hours post-injection was noteworthy. While the DMSO solution itself at high concentration is generally considered toxic, the occurrence of such a recovery in an individual animal may point to inherent variability in the response of mice to the DMSO. It was plausible that the special mouse had a stronger immune or repair response, which allowed it to gradually recover from the toxic effects of the DMSO injection. However, this singular recovery did not diminish the overall trend of severe toxicity observed in the DMSO group, which highlighted the potent and rapid toxicity of DMSO exposure.25

    In mice with injection of the DMSO and MNU combination, the DMSO and MNU combination did not exhibit significantly aggravated toxic reactions or increased mortality in mice during the early stages. From the survival curves, it was apparent that there were no noticeable differences in the early stages compared to the DMSO injection. However, as time progressed, the mortality rate in the DMSO and MNU combination groups began to increase significantly. Upon further analysis, it was found that in the DMSO and MNU combination groups with different doses of MNU, there was no clear MNU-dose dependent increase in toxicity at the early stages. Surprisingly, the early mortality rate in the L group was slightly higher compared to H group. As the observation period extended, however, the mortality rates between the two groups became similar. The results might suggest that the relationship between the MNU dosage and the related toxicity may not always follow a straightforward, dose-dependent pattern. It implied that other factors, such as the rate of absorption, metabolism, or individual genetic responses, might influence the observed outcomes of possible toxicity. The early increased mortality in the L group might be attributed to a higher sensitivity of certain physiological systems or an accelerated onset of toxicity in those mice. As time went on, however, the toxic effects of MNU probably reached a threshold, and the overall mortality rate stabilized in both L and H groups.

    MNU was initially recognized as a potent carcinogenic agent and has been widely used to induce various types of cancer in animal models.26 Subsequent studies demonstrated that MNU at the dose 60 mg/kg could selectively induce apoptosis in retinal photoreceptor cells, which led to its use in research on retinal pigment degeneration and retinal degeneration-related diseases.7,27 However, there has been limited researches on the effects of MNU in increasing animal mortality and lethality. Some studies reported that higher doses of MNU (80, 160 or 240 mg/kg) could lead to tumor formation, leukemia, and even an increase in early mortality in mice26,28. MNU exposure was showed to impair hippocampal neurogenesis in rats,28 which might lead to toxic behaviors. Our study utilized a relatively low dose of MNU, ranging from 40 to 60 mg/kg, and the results suggested that MNU could, to some extent, enhance the toxic effects of DMSO in mice. This enhancement may be due to an increased inherent toxicity of MNU itself, or a result of MNU-mediated potentiation of the DMSO toxicity.

    Several limitations existed in our study. One limitation of this study was the relatively small sample size, which may reduce the statistical power and generalizability of the findings. As our research involved potentially severe toxicological effects and mortality endpoints of mice, we adhered to the principle of minimizing animal use while maintaining scientific validity. This approach aligned with the 3Rs (Replacement, Reduction, and Refinement) principle in animal research. Although we applied the Bootstrap method due to the small sample size, the results should be interpreted with caution and considered as preliminary observations that provided the foundation for more robust studies. Additionally, the short observation period of 48 hours post-injection in our study did not account for potential long-term toxic effects or delayed pathological changes, limiting our understanding of DMSO and MNUinduced chronic toxicity. Besides, the current study did not consider the impact of gender and age variations, which could influence the results. Furthermore, while the study focused on acute toxicity, it did not explore the underlying molecular mechanisms, such as DNA damage repair or oxidative stress. In addition, we did harvest the blood or organs for future enzymatic assays, which might extract the maximum amount of information on the toxicity mechanism. Finally, the use of mice as model organisms, though standard, might not fully represent the human responses. This limitation underscored the necessity for future studies to incorporate other animal models, cell models or human data for broader applicability.

    In conclusion, our study demonstrated that DMSO exhibited a notable toxicity, particularly when used in combination with MNU, the alkylating agents. This combination was shown to significantly reduce the activity levels and increase the mortality rate in mice. These findings highlighted the importance of closely monitoring animal’ reactions during experiments involving DMSO and similar reagents. To mitigate potential adverse effects, it is crucial to establish comprehensive research observation protocols that ensure the health and well-being of the animals. Further studies are needed to explore the underlying mechanisms of DMSO and MNU toxicity and to determine the safest concentrations and guidelines for its application in scientific research. Besides, we would include enzymatic assays of relevant serum and organs, including the heart, liver, spleen, lungs, and kidneys of mice, to further explore the possible toxicity mechanism. By advancing the understanding of DMSO and MNU toxicity, it could better inform safety practices and contribute to the broader field of chemical toxicity assessment.

    Data Sharing Statement

    The datasets used and/or analyzed during the current study were available from the corresponding author on reasonable request.

    Acknowledgments

    We are grateful to our families for their constant support and encouragement throughout this study.

    Weiming Yan, Qiurui He and Lin Xiao are Co-first author.

    Funding

    This work was supported by the grants from the National Natural Science Foundation of China (Grant number: 82301245), the Natural Science Foundation of Fujian Province, China (Grant number: 2024J011148), the Joint Funds for the innovation of science and Technology, Fujian province (Grant number: 2024Y9653), the Postdoctoral Science Foundation of the Fuzhou General Hospital (Grant number: 48678) and the Natural Science Foundation of Zhangzhou City, China (Grant number: ZZ2024J57).

    Disclosure

    The authors declared no potential conflicts of interest regarding the research, authorship, or publication of this article.

    References

    1. Kumar S, Islam R, O’Connor W, et al. A metabolomic analysis on the toxicological effects of the universal solvent, dimethyl sulfoxide. Comp Biochem Physiol C Toxicol Pharmacol. 2025;287:110073. doi:10.1016/j.cbpc.2024.110073

    2. Kligman AM. Dimethyl Sulfoxide. 2. JAMA. 1965;193:923–928. doi:10.1001/jama.1965.03090110061015

    3. Kaczor-Kaminska M, Kaszuba K, Bilska-Wilkosz A, et al. Dimethyl Sulfoxide (DMSO) as a potential source of interference in research related to sulfur metabolism-a preliminary study. Antioxidants. 2024;13(5):582. doi:10.3390/antiox13050582

    4. Sasatani M, Xi Y, Daino K, et al. Rev1 overexpression accelerates N -methyl- N -nitrosourea (MNU)-induced thymic lymphoma by increasing mutagenesis. Cancer Sci. 2024;115(6):1808–1819. doi:10.1111/cas.16159

    5. Kong Q, Han X, Cheng H, et al. Lycium barbarum glycopeptide (wolfberry extract) slows N-methyl-N-nitrosourea-induced degradation of photoreceptors. Neural Regen Res. 2024;19(10):2290–2298. doi:10.4103/1673-5374.390958

    6. Tawarayama H, Uchida K, Hasegawa H, et al. Estrogen, via ESR2 receptor, prevents oxidative stress-induced Muller cell death and stimulates FGF2 production independently of NRF2, attenuating retinal degeneration. Exp Eye Res. 2024;248:110103. doi:10.1016/j.exer.2024.110103

    7. Zhu Y, Zhao X, Japhet TJ, Chen J, Zhao Q. Sampling of N-methyl-N-nitrosourea-induced rat’s retina damage and multi-index evaluation of pathological changes. J Vis Exp. 2024;(211).

    8. Delgado-Goni T, Martín‐Sitjar J, Simões RV, et al. Dimethyl sulfoxide (DMSO) as a potential contrast agent for brain tumors. NMR Biomed. 2013;26(2):173–184. doi:10.1002/nbm.2832

    9. Rosch S, Werner C, Müller F, et al. Photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea (MNU) in rabbits: a pilot study. Graefes Arch Clin Exp Ophthalmol. 2017;255(2):317–331. doi:10.1007/s00417-016-3531-7

    10. Yan W, He Q, Long P, et al. A novelly-spatiotemporal characterization of the disease course in the MNU-induced retinitis pigmentosa model. J Inflamm Res. 2024;17:9243–9254. doi:10.2147/JIR.S474102

    11. Tanaka M, Abe S. Augmented anti-tumour effect of lipopolysaccharide with G-CSF without enhancing body weight loss in mice bearing MH134 hepatoma. Eur J Pharmacol. 2022;934:175206. doi:10.1016/j.ejphar.2022.175206

    12. Zhan J, Ma X, Liu D, et al. Gut microbiome alterations induced by tributyltin exposure are associated with increased body weight, impaired glucose and insulin homeostasis and endocrine disruption in mice. Environ Pollut. 2020;266(Pt 3):115276. doi:10.1016/j.envpol.2020.115276

    13. Jarrar Q, Ayoub R, Jarrar Y, et al. Flumazenil pretreatment reduces mefenamic acid-induced central nervous system toxicity in mice. J Integr Neurosci. 2023;22(4):104. doi:10.31083/j.jin2204104

    14. Mohamad Shalan NAA, Mustapha NM, Mohamed S. Chronic toxicity evaluation of Morinda citrifolia fruit and leaf in mice. Regul Toxicol Pharmacol. 2017;83:46–53. doi:10.1016/j.yrtph.2016.11.022

    15. Nguyen KC, Zhang Y, Todd J, et al. Biodistribution and systemic effects in mice following intravenous administration of cadmium telluride quantum dot nanoparticles. Chem Res Toxicol. 2019;32(8):1491–1503. doi:10.1021/acs.chemrestox.8b00397

    16. Chadalavada D, Adamson TW, Burnett JC, et al. Irradiated compared with nonirradiated NSG mice for the development of a human B-cell lymphoma model. Comp Med. 2014;64(3):179–185.

    17. Suzuki H, Okada Y. Comparative toxicity of dinophysistoxin-1 and okadaic acid in mice. J Vet Med Sci. 2018;80(4):616–619. doi:10.1292/jvms.17-0377

    18. Uttekar PS, Yadav VD, Bhagwat DA. 1, 2-Dihexadecanoyl-sn-glycero-3-phosphoethanolamin (DPPE), doxorubicin and folic acid conjugated micelles for cancer management in tumor bearing BALB/c mice. Bioorg Med Chem Lett. 2021;50:128337. doi:10.1016/j.bmcl.2021.128337

    19. Dwivedi AK, Mallawaarachchi I, Alvarado LA. Analysis of small sample size studies using nonparametric bootstrap test with pooled resampling method. Stat Med. 2017;36(14):2187–2205. doi:10.1002/sim.7263

    20. Neuhauser M, Jockel K. A bootstrap test for the analysis of microarray experiments with a very small number of replications. Appl Bioinformatics. 2006;5(3):173–179. doi:10.2165/00822942-200605030-00005

    21. Swiecilo A, Januś E, Krzepiłko A, et al. The effect of DMSO on Saccharomyces cerevisiae yeast with different energy metabolism and antioxidant status. Sci Rep. 2024;14(1):21974. doi:10.1038/s41598-024-72400-4

    22. Kim K, Lee S. Combined toxicity of dimethyl sulfoxide (DMSO) and vanadium towards zebrafish embryos (Danio rerio): unexpected synergistic effect by DMSO. Chemosphere. 2021;270:129405. doi:10.1016/j.chemosphere.2020.129405

    23. Haschek WM, Baer KE, Rutherford JE. Effects of dimethyl sulfoxide (DMSO) on pulmonary fibrosis in rats and mice. Toxicology. 1989;54(2):197–205. doi:10.1016/0300-483X(89)90045-0

    24. Huang Y, Cartlidge R, Walpitagama M, et al. Unsuitable use of DMSO for assessing behavioral endpoints in aquatic model species. Sci Total Environ. 2018;615:107–114. doi:10.1016/j.scitotenv.2017.09.260

    25. Fry LJ, Querol S, Gomez SG, et al. Assessing the toxic effects of DMSO on cord blood to determine exposure time limits and the optimum concentration for cryopreservation. Vox Sang. 2015;109(2):181–190. doi:10.1111/vox.12267

    26. Da Silva Franchi CA, Bacchi MM, Padovani CR, et al. Thymic lymphomas in Wistar rats exposed to N-methyl-N-nitrosourea (MNU). Cancer Sci. 2003;94(3):240–243. doi:10.1111/j.1349-7006.2003.tb01427.x

    27. Guo D, Sun Y, Wu J, et al. Photoreceptor-targeted extracellular vesicles-mediated delivery of Cul7 siRNA for retinal degeneration therapy. Theranostics. 2024;14(13):4916–4932. doi:10.7150/thno.99484

    28. Song P, Li X, Chen S, et al. YTHDF1 mediates N -methyl- N -nitrosourea-induced gastric carcinogenesis by controlling HSPH1 translation. Cell Prolif. 2024;57(7):e13619. doi:10.1111/cpr.13619

    Continue Reading

  • Initial community response to a novel spatial repellent for malaria prevention in Busia County, Kenya | Malaria Journal

    Initial community response to a novel spatial repellent for malaria prevention in Busia County, Kenya | Malaria Journal

    A total of 60 interviews were completed during the first two rounds of data collection, covering 30 households in each round. The participants included 16 women and 14 men. The mean age of the participants was 49 years (range 35–65), with women averaging 44 years (range 35–55) and men 55 years (range 45–65).

    In addition to commenting on perceived efficacy, perceived reduction in malaria cases, and side effects, participants compared the perceived performance of SR with other mosquito control methods they commonly used such as bednets, mosquito coils and burning leaves. The participants also discussed their communication with others about the product and suggested future improvements.

    Perceived efficacy and early acceptability

    In interviews conducted one week after initial installation, most participants reported that they noticed a reduction in mosquito density and attributed this reduction to the SR’s dawa [the power or strength of the insecticide]. Those who observed mosquitoes in the house reported that they appeared sluggish, biting and flying less aggressively than they did in the past.

    Generally, since you installed these products inside here, I have seen the mosquito population completely reduced. I can say by 99%. Previously, approximately around 7:00 p.m. while seated here [pointing at the sofa set], we would be bitten by mosquitos until when we went to sleep at 9:00 pm. Now, after the installation of the SR, the few mosquitos we see seem sluggish, often flying slowly, and even falling on their own. Additionally, since the installation, I haven’t heard any child complaining about feeling sick.” (Male, age 39, 1-week post-installation).

    In the second interview, 2 months after the initial installation, some participants reported that mosquitoes had returned. They attributed this return to the belief that replacement SRs had less dawa than the initial product and lost their effectiveness after 2–3 weeks.

    How come we are now seeing signs of mosquitos, yet we were praising this product? We are requesting they improve in putting a lot of repellent so that mosquitos do not appear and later disappear. I want, if the product has been installed to repel mosquitos, let it repel mosquitos.” (Male, age 41, 2-month post-installation).

    Some participants also mentioned seasonality, suggesting that the increase in mosquito frequency coincided with millet flowering or the decrease in recent hot weather. One proposed that installing the product during a peak mosquito season could have offered a clearer assessment of its effectiveness.

    While most of the comments focused on mosquito density, some respondents added that fewer family members, particularly children, were falling ill with malaria.

    Since these products were installed in our houses, I have noticed that the number of mosquitos have decreased, and the level of malaria has also decreased. Because before installation, my youngest child had malaria every month, she used to be admitted to the ward. However, since these products were installed, she has never has never been sick.” (Female, age 28, 2-month post-installation).

    In addition to reporting perceived reductions in mosquitoes and malaria symptoms, many participants expressed satisfaction with the SR. They described it as convenient, easy to use, and preferable to other mosquito control tools like nets or coils. These early expressions of acceptance suggest a positive initial reception of the product.

    Comparison of SRs to other mosquito control products

    The participants reported using other products and practices to keep away mosquitoes, including ITNs, mosquito coils, mosquito mats, and burning leaves to produce smoke that repels mosquitoes.

    When asked to compare the SR to other products, respondents cited various factors including the place of protection, the cost, the product’s perceived effectiveness, and the feasibility of installation. Some mentioned that ITNs only protected them while they were sleeping whereas the SRs also offered protection when they were awake. Others reported that SRs obviated the need for ITNs.

    I can say that the SR is better than a net because a net is only used when you go to sleep. You only protect yourself with it during sleep. But with the SR, you can be protected while sitting in the evening. Mosquitoes won’t bother me because of the SR. However, with a net, you must wait until you go to sleep at 10:00 p.m to be protected.” (Female, age 32, 1-week post-installation).

    Because they have installed a product for me that repels mosquitos, I do not see the need to struggle hanging the net because it brings heat. You know during dry season like this there is a lot of heat. There is no need to interfere with the product that repels mosquitos, why not sleep comfortably?” (Male, age 41, 2-month post-installation).

    The respondents also stated that the SR was less labour-intensive than an ITN and that ITNs could tear, make their living spaces hotter, and cause irritation for those who came into direct contact with them.

    It’s our first time to use spatial repellant, and we are still observing the effects. But within this short time, I see it’s effective and it’s chasing away the mosquitoes even while inside the net, the treated net may lose its effectiveness with time, then the mosquitoes just gain access to you.” (Male, age 63, 1-week post-installation).

    Some added that SRs improved household finances since the study provided them at no cost.

    The spatial repellent is good because since they were installed in my house, I do not use any money by going to buy other mosquito repellents. But when I did not have [SR], mosquito coils used to cost me money. When you budget for supper, you must put the mosquito coil budget there too.” (Female, age 28, 1-week post-installation).

    Perceived side effects

    While most respondents reported no side effects within their own households, some mentioned observing effects on non-target organisms, including insects such as cockroaches and small animals. One participant described seeing cockroaches that appeared weakened or dead after installation of the SR:

    What I have experienced with the cockroaches in this house, you find them moving and are weak and some are even dead and others are unconscious there. They are not moving as they used to before and hiding in private places. Currently this product is also affecting them too.” (Female, age 38, 2-month post installation).

    Many viewed these effects as an additional benefit of SRs. Some, however, cited conversations in which a neighbor had mentioned side effects such as children sneezing when they got too close to the product or skin irritation experienced by one participant’s husband after touching it. Others expressed uncertainty about whether the effects experienced since installation were directly caused by the SR or by other factors. As a precaution, some participants reported keeping children at a distance from the product to avoid potential adverse effects. Those ‘Ikee’ [Ateso word for medicine or active ingredient, referring to the SR] you are not supposed to get close. At times when you are putting things in order, you need to keep distance from the ‘Ikee’ because we were advised that way. There was a time when my brother’s child who stays here, he got closer to the ‘Ikee’, he started sneezing and I told him that he is not allowed to be close.” (Female, age 41, 2-month post installation).

    Suggestions for future improvement

    At the first TIPs visit, the participants suggested few improvements to the SR, possibly due to their limited experience. Subsequently informants suggested changes in size, shape, colour, smell, installation method, and replacement frequency. A participant suggested alternating the SR’s colour with each replacement not due to aesthetic preference, but to make it easier to recognize whether the product has been changed especially when residents were away during installation.

    Other mentioned that white was best because it was visible and matched house décor, while a few said colour did not matter as long as the product worked. Regarding shape and size, some participants suggested enlarging SRs could reduce the number of units required per household. They said they liked that the product was odourless and its general appearance but recommended extending installations to areas such as bathrooms, latrines, and schools where mosquito encounters were common, particularly during the early morning.

    Then secondly, there are those places these products were not installed. Places like the toilet. If you can find those, then you install. You know you go there anytime, it is dark, and you get bitten. Then again, within here in our home, children study in school. Our children always leave early. Exactly by 6:00 a.m. they are in class. They can get bitten by mosquitoes while there. If it is possible, they should take [SRs] to schools too.” (Male, 39, 2-month post-installation).

    The participants expressed concerns about the product falling off the wall when installed with tape, which led one family to rehang a fallen product with a nail. They stated their preference for hooks, which kept SRs more securely attached to the wall (Figs. 2 and 3). Some suggested offering a range of colours (black, white, green, blue, khaki) for the products, with one linking a dark colour to attracting mosquitoes.

    The majority expressed a preference for an odorless product, considering allergies, but a few suggested that some scent was necessary for effective mosquito repellency. A recurring recommendation was to increase the amount or concentration of repellent, with diverse opinions on replacement intervals. Some participants suggested a switch to biodegradable materials to address the environmental concerns associated with plastic sheets.Another one is, we were also saying, they should try and look for a product that is friendly to the environment apart from these papers. At least, even if the project ends and they left the products for us they have not come to collect. At least it should be a material that when you throw, it rots faster.” (Male, age 39, 2-month post installation).

    Fig. 2

    Family rehung fallen taped product with nail

    Fig. 3
    figure 3

    Installation of the MosquitoShield™ with hooks

    Continue Reading

  • KSRelief launches 3rd cycle of food security support project in Pakistan – RADIO PAKISTAN

    1. KSRelief launches 3rd cycle of food security support project in Pakistan  RADIO PAKISTAN
    2. Saudi envoy launches food aid drive, reaffirms Kingdom’s unwavering support for Pakistan  Associated Press of Pakistan
    3. KSrelief launches third cycle of food security support project in Pakistan  Associated Press of Pakistan

    Continue Reading

  • WHO. World malaria report 2023. Geneva: World Health Organization; 2023. Available at: https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2023. Accessed April 2024.

  • WHO. World malaria report 2024: addressing inequity in the global malaria response. Geneva: World Health Organization; 2024. Available at: https://www.who.int/teams/global-malaria-programme/reports/world-malaria-report-2024. Accessed January 2025.

  • Singh B, Kim Sung L, Matusop A, Radhakrishnan A, Shamsul SS, Cox-Singh J, et al. A large focus of naturally acquired Plasmodium knowlesi infections in human beings. Lancet. 2004;363:1017–24.

    Google Scholar 

  • Jongwutiwes S, Putaporntip C, Iwasaki T, Sata T, Kanbara H. Naturally acquired Plasmodium knowlesi malaria in human, Thailand. Emerg Infect Dis. 2004;10:2211–3.

    PubMed Central 

    Google Scholar 

  • Zhu HM, Li J, Zheng H. Human natural infection of Plasmodium knowlesi. Zhongguo Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi. 2006;24:70–1.

    Google Scholar 

  • Khim N, Siv S, Kim S, Mueller T, Fleischmann E, Singh B, et al. Plasmodium knowlesi infection in humans, Cambodia, 2007–2010. Emerg Infect Dis. 2011;17:1900–2.

    PubMed Central 

    Google Scholar 

  • Van den Eede P, Van HN, Van Overmeir C, Vythilingam I, Duc TN, le Hung X, et al. Human Plasmodium knowlesi infections in young children in central Vietnam. Malar J. 2009;8:249.

    PubMed Central 

    Google Scholar 

  • Iwagami M, Nakatsu M, Khattignavong P, Soundala P, Lorphachan L, Keomalaphet S, et al. First case of human infection with Plasmodium knowlesi in Laos. PLoS Negl Trop Dis. 2018;12: e0006244.

    PubMed Central 

    Google Scholar 

  • WHO. The Mekong Malaria Elimination programme. Accelerating malaria elimination in the Greater Mekong. Bulletin 10, March 2022. Geneva: World Health Organization; 2022. Available at: https://www.who.int/publications/i/item/WHO-UCN-GMP-MME-2022.01. Accessed August 2024.

  • Zaw AS, Win ESS, Yan SW, Thein KS, Verma V, McLean ARD, et al. Successful elimination of falciparum malaria following the introduction of community-based health workers in Eastern Myanmar: a retrospective analysis. PLoS Med. 2023;20: e1004318.

    PubMed Central 

    Google Scholar 

  • Tun STT, Von Seidlein L, Pongvongsa T, Mayxay M, Saralamba S, Kyaw SS, et al. Towards malaria elimination in Savannakhet, Lao PDR: mathematical modelling driven strategy design. Malar J. 2017;16:1–12.

    Google Scholar 

  • WHO. Malaria Policy Advisory Group (MPAG) meeting, March 2022. Geneva: World Health Organization; 2022. Available at: https://www.who.int/publications/i/item/9789240048430. Accessed January 2025.

  • Johnson E, Sunil Kumar Sharma R, Ruiz Cuenca P, Byrne I, Salgado-Lynn M, Suraya Shahar Z, et al. Landscape drives zoonotic malaria prevalence in non-human primates. Elife. 2024;12: RP88616.

  • Woolley SD, Beeching NJ, Lalloo DG, Rajahram GS. Is the rise of simian zoonotic malarias a public health problem caused by humans? A review of simian malaria in humans. IJID One Health. 2023;1: 100002.

    Google Scholar 

  • Hartmeyer GN, Stensvold CR, Fabricius T, Marmolin ES, Hoegh SV, Nielsen HV, et al. Plasmodium cynomolgi as cause of malaria in tourist to Southeast Asia, 2018. Emerg Infect Dis. 2019;25:1936–9.

    PubMed Central 

    Google Scholar 

  • Fornace KM, Zorello Laporta G, Vythilingham I, Chua TH, Ahmed K, Jeyaprakasam NK, et al. Simian malaria: a narrative review on emergence, epidemiology and threat to global malaria elimination. Lancet Infect Dis. 2023;23:e520–32.

    Google Scholar 

  • Hii J, Vythilingam I, Roca-Feltrer A. Human and simian malaria in the Greater Mekong Subregion and challenges for elimination. Rijeka: InTechOpen. 2018;2018:95–127.

  • Ngassa Mbenda HG, Wang M, Guo J, Siddiqui FA, Hu Y, Yang Z, et al. Evolution of the Plasmodium vivax multidrug resistance 1 gene in the Greater Mekong Subregion during malaria elimination. Parasit Vectors. 2020;13:67.

    CAS 
    PubMed Central 

    Google Scholar 

  • Chareonviriyaphap T, Bangs MJ, Suwonkerd W, Kongmee M, Corbel V, Ngoen-Klan R. Review of insecticide resistance and behavioral avoidance of vectors of human diseases in Thailand. Parasit Vectors. 2013;6:1–28.

    Google Scholar 

  • Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. 2021;372: n160.

    PubMed Central 

    Google Scholar 

  • Munn Z, Stern C, Aromataris E, Lockwood C, Jordan Z. What kind of systematic review should I conduct? A proposed typology and guidance for systematic reviewers in the medical and health sciences. BMC Med Res Methodol. 2018;18:5.

    PubMed Central 

    Google Scholar 

  • Munn Z, Moola S, Lisy K, Riitano D, Tufanaru C. Methodological guidance for systematic reviews of observational epidemiological studies reporting prevalence and cumulative incidence data. JBI Evid Implement. 2015;13:147–53.

    Google Scholar 

  • Barendregt JJ, Doi SA, Lee YY, Norman RE, Vos T. Meta-analysis of prevalence. J Epidemiol Community Health. 2013;67:974–8.

  • RStudio Team. RStudio: Integrated Development Environment for R. Version 2024.09.1+394 [software]. Posit Software, PBC; 2024. Available from: https://posit.co. Accessed June 2024.

  • Viechtbauer W. Conducting meta-analyses in R with the metafor package. J Stat Softw. 2010;36:1–48.

    Google Scholar 

  • Hadley W. ggplot2: elegant graphics for data analysis. Springer Cham; 2016.

  • Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002;21:1539–58.

    Google Scholar 

  • Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315:629–34.

    CAS 
    PubMed Central 

    Google Scholar 

  • Jongwutiwes S, Buppan P, Kosuvin R, Seethamchai S, Pattanawong U, Sirichaisinthop J, et al. Plasmodium knowlesi malaria in humans and macaques, Thailand. Emerg Infect Dis. 2011;17:1799–806.

    PubMed Central 

    Google Scholar 

  • Putaporntip C, Hongsrimuang T, Seethamchai S, Kobasa T, Limkittikul K, Cui L, et al. Differential prevalence of Plasmodium infections and cryptic Plasmodium knowlesi malaria in humans in Thailand. J Infect Dis. 2009;199:1143–50.

    CAS 

    Google Scholar 

  • Putaporntip C, Kuamsab N, Pattanawong U, Yanmanee S, Seethamchai S, Jongwutiwes S. Plasmodium cynomolgi co-infections among symptomatic malaria patients, Thailand. Emerg Infect Dis. 2021;27:590–3.

    PubMed Central 

    Google Scholar 

  • Sermwittayawong N, Singh B, Nishibuchi M, Sawangjaroen N, Vuddhakul V. Human Plasmodium knowlesi infection in Ranong province, southwestern border of Thailand. Malar J. 2012;11:36.

    PubMed Central 

    Google Scholar 

  • Marchand RP, Culleton R, Maeno Y, Quang NT, Nakazawa S. Co-infections of Plasmodium knowlesi, P. falciparum, and P. vivax among humans and Anopheles dirus mosquitoes, Southern Vietnam. Emerg Infect Dis. 2011;17:1232–9.

  • Shimizu S, Chotirat S, Dokkulab N, Hongchad I, Khowsroy K, Kiattibutr K, et al. Malaria cross-sectional surveys identified asymptomatic infections of Plasmodium falciparum, Plasmodium vivax and Plasmodium knowlesi in Surat Thani, a southern province of Thailand. Int J Infect Dis. 2020;96:445–51.

    CAS 

    Google Scholar 

  • Sugaram R, Boondej P, Srisutham S, Kunasol C, Pagornrat W, Boonyuen U, et al. Genetic population of Plasmodium knowlesi during pre-malaria elimination in Thailand. Malar J. 2021;20:454.

    CAS 
    PubMed Central 

    Google Scholar 

  • Ghinai I, Cook J, Hla TT, Htet HM, Hall T, Lubis IN, et al. Malaria epidemiology in central Myanmar: identification of a multi-species asymptomatic reservoir of infection. Malar J. 2017;16:16.

    PubMed Central 

    Google Scholar 

  • Imwong M, Madmanee W, Suwannasin K, Kunasol C, Peto TJ, Tripura R, et al. Asymptomatic Natural Human Infections With the Simian Malaria Parasites Plasmodium cynomolgi and Plasmodium knowlesi. J Infect Dis. 2019;219:695–702.

    CAS 

    Google Scholar 

  • Jiang N, Chang Q, Sun X, Lu H, Yin J, Zhang Z, et al. Co-infections with Plasmodium knowlesi and other malaria parasites, Myanmar. Emerg Infect Dis. 2010;16:1476–8.

    PubMed Central 

    Google Scholar 

  • Yek C, Lay S, Bohl JA, Man S, Chea S, Lon C, et al. Case Report: cambodian national malaria surveillance program detection of Plasmodium knowlesi. Am J Trop Med Hyg. 2022;107:151–3.

    PubMed Central 

    Google Scholar 

  • Barber BE, William T, Dhararaj P, Anderios F, Grigg MJ, Yeo TW, et al. Epidemiology of Plasmodium knowlesi malaria in north-east Sabah, Malaysia: family clusters and wide age distribution. Malar J. 2012;11:1–8.

    Google Scholar 

  • Barber BE, William T, Grigg MJ, Menon J, Auburn S, Marfurt J, et al. A prospective comparative study of knowlesi, falciparum, and vivax malaria in Sabah, Malaysia: high proportion with severe disease from Plasmodium knowlesi and Plasmodium vivax but no mortality with early referral and artesunate therapy. Clin Infect Dis. 2012;56:383–97.

    Google Scholar 

  • Barber BE, William T, Jikal M, Jilip J, Dhararaj P, Menon J, et al. Plasmodium knowlesi malaria in children. Emerg Infect Dis. 2011;17:814–20.

    PubMed Central 

    Google Scholar 

  • Chin AZ, Avoi R, Atil A, Lukman KA, Rahim SSSA, Ibrahim MY, et al. Risk factor of Plasmodium knowlesi infection in Sabah Borneo Malaysia, 2020: a population-based case-control study. PLoS ONE. 2021;16: e0257104.

    CAS 
    PubMed Central 

    Google Scholar 

  • Cooper DJ, Rajahram GS, William T, Jelip J, Mohammad R, Benedict J, et al. Plasmodium knowlesi Malaria in Sabah, Malaysia, 2015–2017: ongoing increase in incidence despite near elimination of the human-only Plasmodium species. Clin Infect Dis. 2020;70:361–7.

    Google Scholar 

  • Daneshvar C, Davis TM, Cox-Singh J, Rafa’ee MZ, Zakaria SK, Divis PC, et al. Clinical and laboratory features of human Plasmodium knowlesi infection. Clin Infect Dis. 2009;49:852–60.

    Google Scholar 

  • Dian ND, Muhammad AB, Azman EN, Eddie NA, Azmi NI, Yee VCT, et al. Evidence of submicroscopic Plasmodium knowlesi mono-infection in remote indigenous communities in Kelantan, Peninsular Malaysia. Am J Trop Med Hyg. 2023;109:1081–5.

    CAS 
    PubMed Central 

    Google Scholar 

  • Fornace KM, Abidin TR, Alexander N, Brock P, Grigg MJ, Murphy A, et al. Association between landscape factors and spatial patterns of Plasmodium knowlesi infections in Sabah, Malaysia. Emerg Infect Dis. 2016;22:201–8.

    CAS 
    PubMed Central 

    Google Scholar 

  • Fornace KM, Herman LS, Abidin TR, Chua TH, Daim S, Lorenzo PJ, et al. Exposure and infection to Plasmodium knowlesi in case study communities in Northern Sabah, Malaysia and Palawan, The Philippines. PLoS Negl Trop Dis. 2018;12: e0006432.

    PubMed Central 

    Google Scholar 

  • Goh XT, Lim YA, Vythilingam I, Chew CH, Lee PC, Ngui R, et al. Increased detection of Plasmodium knowlesi in Sandakan division, Sabah as revealed by PlasmoNex™. Malar J. 2013;12:264.

    PubMed Central 

    Google Scholar 

  • Grigg MJ, Cox J, William T, Jelip J, Fornace KM, Brock PM, et al. Individual-level factors associated with the risk of acquiring human Plasmodium knowlesi malaria in Malaysia: a case-control study. Lancet Planet Health. 2017;1:e97–104.

    PubMed Central 

    Google Scholar 

  • Grignard L, Shah S, Chua TH, William T, Drakeley CJ, Fornace KM. Natural human infections with Plasmodium cynomolgi and other malaria species in an elimination setting in Sabah, Malaysia. J Infect Dis. 2019;220:1946–9.

    PubMed Central 

    Google Scholar 

  • Jiram AI, Hisam S, Reuben H, Husin SZ, Roslan A, Wan Ismail WR. Submicroscopic evidence of the simian malaria parasite, Plasmodium knowlesi, in an Orang Asli community, Southeast Asian. J Trop Med and Public Health. 2016;47:591–9.

    Google Scholar 

  • Jiram AI, Ooi CH, Rubio JM, Hisam S, Karnan G, Sukor NM, et al. Evidence of asymptomatic submicroscopic malaria in low transmission areas in Belaga district, Kapit division, Sarawak, Malaysia. Malar J. 2019;18:156.

    PubMed Central 

    Google Scholar 

  • Joveen-Neoh WF, Chong KL, Wong CM, Lau TY. Incidence of malaria in the interior division of Sabah, Malaysian Borneo, based on nested PCR. J Parasitol Res. 2011;2011: 104284.

    PubMed Central 

    Google Scholar 

  • Lai MY, Rafieqin N, Lee PYL, Amir Rawa MS, Dzul S, Yahaya N, et al. High incidence of Plasmodium knowlesi malaria compared to other human malaria species in several hospitals in Malaysia. Trop Biomed. 2021;38:248–53.

    CAS 

    Google Scholar 

  • Lee KS, Cox-Singh J, Brooke G, Matusop A, Singh B. Plasmodium knowlesi from archival blood films: further evidence that human infections are widely distributed and not newly emergent in Malaysian Borneo. Int J Parasitol. 2009;39:1125–8.

    PubMed Central 

    Google Scholar 

  • Lee PC, Chong ET, Anderios F, Al Lim Y, Chew CH, Chua KH. Molecular detection of human Plasmodium species in Sabah using PlasmoNex™ multiplex PCR and hydrolysis probes real-time PCR. Malar J. 2015;14:28.

    PubMed Central 

    Google Scholar 

  • Noordin NR, Lee PY, Mohd Bukhari FD, Fong MY, Abdul Hamid MH, Jelip J, et al. Prevalence of asymptomatic and/or low-density malaria infection among high-risk groups in Peninsular Malaysia. Am J Trop Med Hyg. 2020;103:1107–10.

    CAS 
    PubMed Central 

    Google Scholar 

  • Pramasivan S, Ngui R, Jeyaprakasam NK, Liew JWK, Low VL, Mohamed Hassan N, et al. Spatial distribution of Plasmodium knowlesi cases and their vectors in Johor, Malaysia: in light of human malaria elimination. Malar J. 2021;20:426.

    CAS 
    PubMed Central 

    Google Scholar 

  • Siner A, Liew ST, Kadir KA, Mohamad DSA, Thomas FK, Zulkarnaen M, et al. Absence of Plasmodium inui and Plasmodium cynomolgi, but detection of Plasmodium knowlesi and Plasmodium vivax infections in asymptomatic humans in the Betong division of Sarawak, Malaysian Borneo. Malar J. 2017;16:417.

    PubMed Central 

    Google Scholar 

  • William T, Jelip J, Menon J, Anderios F, Mohammad R, Awang Mohammad TA, et al. Changing epidemiology of malaria in Sabah, Malaysia: increasing incidence of Plasmodium knowlesi. Malar J. 2014;13:390.

    PubMed Central 

    Google Scholar 

  • Yap NJ, Hossain H, Nada-Raja T, Ngui R, Muslim A, Hoh BP, et al. Natural human Infections with Plasmodium cynomolgi, P. inui, and 4 other simian malaria parasites, Malaysia. Emerg Infect Dis. 2021;27:2187–91.

  • Yunos NE, Sharkawi HM, Hii KC, Hu TH, Mohamad DSA, Rosli N, et al. Spatio-temporal distribution and hotspots of Plasmodium knowlesi infections in Sarawak, Malaysian Borneo. Sci Rep. 2022;12:17284.

    CAS 
    PubMed Central 

    Google Scholar 

  • Yusof R, Lau YL, Mahmud R, Fong MY, Jelip J, Ngian HU, et al. High proportion of knowlesi malaria in recent malaria cases in Malaysia. Malar J. 2014;13:168.

    PubMed Central 

    Google Scholar 

  • Raja TN, Hu TH, Kadir KA, Mohamad DSA, Rosli N, Wong LL, et al. Naturally acquired human Plasmodium cynomolgi and P. knowlesi infections, Malaysian Borneo. Emerg Infect Dis. 2020;26:1801–9.

  • Pongvongsa T, Culleton R, Ha H, Thanh L, Phongmany P, Marchand RP, et al. Human infection with Plasmodium knowlesi on the Laos-Vietnam border. Trop Med Health. 2018;46:33.

    PubMed Central 

    Google Scholar 

  • Akter R, Vythilingam I, Khaw LT, Qvist R, Lim YAL, Sitam FT, et al. Simian malaria in wild macaques: first report from Hulu Selangor district, Selangor, Malaysia. Malar J. 2015;14:386.

    PubMed Central 

    Google Scholar 

  • Amir A, Shahari S, Liew JWK, de Silva JR, Khan MB, Lai MY, et al. Natural Plasmodium infection in wild macaques of three states in peninsular Malaysia. Acta Trop. 2020;211: 105596.

    CAS 

    Google Scholar 

  • Fungfuang W, Udom C, Tongthainan D, Kadir KA, Singh B. Malaria parasites in macaques in Thailand: stump-tailed macaques (Macaca arctoides) are new natural hosts for Plasmodium knowlesi, Plasmodium inui, Plasmodium coatneyi and Plasmodium fieldi. Malar J. 2020;19:350.

    PubMed Central 

    Google Scholar 

  • Karnchaisri K, Day NPJ, Dondorp AM, Malaivijitnond S, Imwong M. Prevalence and genetic diversity of simian malaria in wild macaque populations across Thailand: implications for human health. Acta Trop. 2024;254: 107187.

    CAS 

    Google Scholar 

  • Lee KS, Divis PC, Zakaria SK, Matusop A, Julin RA, Conway DJ, et al. Plasmodium knowlesi: reservoir hosts and tracking the emergence in humans and macaques. PLoS Pathog. 2011;7: e1002015.

    CAS 
    PubMed Central 

    Google Scholar 

  • Nada-Raja T, Kadir KA, Divis PCS, Mohamad DSA, Matusop A, Singh B. Macaca fascicularis and Macaca nemestrina infected with zoonotic malaria parasites are widely distributed in Sarawak, Malaysian Borneo. Sci Rep. 2022;12:10476.

    CAS 
    PubMed Central 

    Google Scholar 

  • Putaporntip C, Jongwutiwes S, Thongaree S, Seethamchai S, Grynberg P, Hughes AL. Ecology of malaria parasites infecting Southeast Asian macaques: evidence from cytochrome b sequences. Mol Ecol. 2010;19:3466–76.

    CAS 
    PubMed Central 

    Google Scholar 

  • Seethamchai S, Putaporntip C, Malaivijitnond S, Cui L, Jongwutiwes S. Malaria and Hepatocystis species in wild macaques, southern Thailand. Am J Trop Med Hyg. 2008;78:646–53.

    CAS 

    Google Scholar 

  • Shahari S, Bin Abdullah ML, Binti Isman Rohimly AA, Binti Ashrat N, Amir A, Atroosh WMM, et al. The prevalence of simian malaria in wild long-tailed macaques throughout Peninsular Malaysia. Sci Rep. 2024;14:6023.

  • Yusuf NM, Zulkefli J, Jiram AI, Vythilingam I, Hisam S, Devi R, et al. Plasmodium spp. in macaques, Macaca fascicularis, in Malaysia, and their potential role in zoonotic malaria transmission. Parasite. 2022;29:32.

  • Zhang X, Kadir KA, Quintanilla-Zariñan LF, Villano J, Houghton P, Du H, et al. Distribution and prevalence of malaria parasites among long-tailed macaques (Macaca fascicularis) in regional populations across Southeast Asia. Malar J. 2016;15:450.

    PubMed Central 

    Google Scholar 

  • Narapakdeesakul D, Pengsakul T, Kaewparuehaschai M, Thongsahuan S, Moonmake S, Lekcharoen P, et al. Zoonotic simian malaria parasites in free-ranging Macaca fascicularis macaques and human malaria patients in Thailand, with a note on genetic characterization of recent isolates. Acta Trop. 2023;248: 107030.

    CAS 

    Google Scholar 

  • Putaporntip C, Kuamsab N, Seethamchai S, Pattanawong U, Rojrung R, Yanmanee S, et al. Cryptic Plasmodium inui and Plasmodium fieldi infections among symptomatic malaria patients in Thailand. Clin Infect Dis. 2022;75:805–12.

    CAS 

    Google Scholar 

  • Saleh Huddin A, Md Yusuf N, Razak M, Ogu Salim N, Hisam S. Genetic diversity of Plasmodium knowlesi among human and long-tailed macaque populations in Peninsular Malaysia: the utility of microsatellite markers. Infect Genet Evol. 2019;75: 103952.

    Google Scholar 

  • Vythilingam I, Noorazian YM, Huat TC, Jiram AI, Yusri YM, Azahari AH, et al. Plasmodium knowlesi in humans, macaques and mosquitoes in Peninsular Malaysia. Parasit Vectors. 2008;1:26.

    PubMed Central 

    Google Scholar 

  • Judson SD, Rabinowitz PM. Zoonoses and global epidemics. Curr Opin Infect Dis. 2021;34:385–92.

    CAS 

    Google Scholar 

  • Chin W, Contacos PG, Coatney GR, Kimball HR. A naturally acquired quotidian-type malaria in man transferable to monkeys. Science. 1965;149:865.

    CAS 

    Google Scholar 

  • Carneiro I, Roca-Feltrer A, Griffin J, Smith L, Tanner M, Schellenberg J, et al. Age-patterns of malaria vary with severity, transmission intensity and seasonality in Sub-Saharan Africa: a systematic review and pooled analysis. PLoS ONE. 2010;5: e8988.

    PubMed Central 

    Google Scholar 

  • Randall LM, Kenangalem E, Lampah DA, Tjitra E, Mwaikambo ED, Handojo T, et al. Age-related susceptibility to severe malaria associated with galectin-2 in highland Papuans. J Infect Dis. 2010;202:117–24.

    Google Scholar 

  • Davidson G, Chua TH, Cook A, Speldewinde P, Weinstein P. Defining the ecological and evolutionary drivers of Plasmodium knowlesi transmission within a multi-scale framework. Malar J. 2019;18:66.

    PubMed Central 

    Google Scholar 

  • Gallalee S, Zarlinda I, Silaen MG, Cotter C, Cueto C, Elyazar IRF, et al. Forest-goers as a heterogeneous population at high-risk for malaria: a case–control study in Aceh Province, Indonesia. Malar J. 2024;23:37.

    PubMed Central 

    Google Scholar 

  • Fornace KM, Brock PM, Abidin TR, Grignard L, Herman LS, Chua TH, et al. Environmental risk factors and exposure to the zoonotic malaria parasite Plasmodium knowlesi across northern Sabah, Malaysia: a population-based cross-sectional survey. Lancet Planet Health. 2019;3:e179–86.

    PubMed Central 

    Google Scholar 

  • Maeno Y. Molecular epidemiology of mosquitoes for the transmission of forest malaria in south-central Vietnam. Trop Med Health. 2017;45:27.

    PubMed Central 

    Google Scholar 

  • van de Straat B, Sebayang B, Grigg MJ, Staunton K, Garjito TA, Vythilingam I, et al. Zoonotic malaria transmission and land use change in Southeast Asia: what is known about the vectors. Malar J. 2022;21:109.

    PubMed Central 

    Google Scholar 

  • Davidson G, Chua TH, Cook A, Speldewinde P, Weinstein P. The role of ecological linkage mechanisms in Plasmodium knowlesi transmission and spread. EcoHealth. 2019;16:594–610.

    Google Scholar 

  • Iv S, Nguon C, Kong P, Sieng T, Srun S, Christiansen-Jucht C, et al. Intermittent preventive treatment for forest goers by forest malaria workers: an observational study on a key intervention for malaria elimination in Cambodia. Lancet Reg Health West Pac. 2024;47: 101093.

    PubMed Central 

    Google Scholar 

  • WHO. Approaches for mobile and migrant populations in the context of malaria multi-drug resistance and malaria elimination in the Greater Mekong Subregion. Regional Office for South-East Asia, World Health Organization. New Delhi, India; 2016. Available at: http://www.who.int/iris/handle/10665/204351. Accessed August 2024.

  • Bisanzio D, Sudathip P, Kitchakarn S, Kanjanasuwan J, Gopinath D, Pinyajeerapat N, et al. Malaria stratification mapping in Thailand to support prevention of reestablishment. Am J Trop Med Hyg. 2024;110:79–82.

    Google Scholar 

  • BVBD: Bureau of Vectorborne Diseases: Guide to malaria elimination for Thailand’s local administrative organizations and the health network. Department of Disease Control, Ministry of Public Health Bangkok, Thailand; 2019.

  • Sudathip P, Naowarat S, Kitchakarn S, Gopinath D, Bisanzio D, Pinyajeerapat N, et al. Assessing Thailand’s 1-3-7 surveillance strategy in accelerating malaria elimination. Malar J. 2022;21:222.

    PubMed Central 

    Google Scholar 

  • White NJ. Plasmodium knowlesi: the fifth human malaria parasite. Clin Infect Dis. 2008;46:172–3.

    CAS 

    Google Scholar 

  • Tobin RJ, Harrison LE, Tully MK, Lubis IND, Noviyanti R, Anstey NM, et al. Updating estimates of Plasmodium knowlesi malaria risk in response to changing land use patterns across Southeast Asia. PLoS Negl Trop Dis. 2024;18: e0011570.

    PubMed Central 

    Google Scholar 

  • Anstey NM, Grigg MJ, Rajahram GS, Cooper DJ, William T, Kho S, et al. Knowlesi malaria: human risk factors, clinical spectrum, and pathophysiology. Adv Parasitol. 2021;113:1–43.

    PubMed Central 

    Google Scholar 

  • Seilmaier M, Hartmann W, Beissner M, Fenzl T, Haller C, Guggemos W, et al. Severe Plasmodium knowlesi infection with multi-organ failure imported to Germany from Thailand/Myanmar. Malar J. 2014;13:422.

    PubMed Central 

    Google Scholar 

  • William T, Menon J, Rajahram G, Chan L, Ma G, Donaldson S, et al. Severe Plasmodium knowlesi malaria in a tertiary care hospital, Sabah, Malaysia. Emerg Infect Dis. 2011;17:1248–55.

    PubMed Central 

    Google Scholar 

  • Rajahram GS, Cooper DJ, William T, Grigg MJ, Anstey NM, Barber BE. Deaths from Plasmodium knowlesi malaria: case series and systematic review. Clin Infect Dis. 2019;69:1703–11.

    PubMed Central 

    Google Scholar 

  • Ahmed MA, Deshmukh GY, Zaidi RH, Saif A, Alshahrani MA, Wazid SW, et al. Identification, mapping, and genetic diversity of novel conserved cross-species epitopes of RhopH2 in Plasmodium knowlesi with Plasmodium vivax. Front Cell Infect Microbiol. 2021;11: 810398.

    CAS 

    Google Scholar 

  • Manzoni G, Try R, Guintran JO, Christiansen-Jucht C, Jacoby E, Sovannaroth S, et al. Progress towards malaria elimination in the Greater Mekong Subregion: perspectives from the World Health Organization. Malar J. 2024;23:64.

    PubMed Central 

    Google Scholar 

  • Mahittikorn A, Masangkay FR, Kotepui KU, Milanez GJ, Kotepui M. Quantification of the misidentification of Plasmodium knowlesi as Plasmodium malariae by microscopy: an analysis of 1569 P. knowlesi cases. Malar J. 2021;20:179.

  • Grigg MJ, Lubis IN, Tetteh KKA, Barber BE, William T, Rajahram GS, et al. Plasmodium knowlesi detection methods for human infections-diagnosis and surveillance. Adv Parasitol. 2021;113:77–130.

    PubMed Central 

    Google Scholar 

  • Sulistyaningsih E, Fitri LE, Löscher T, Berens-Riha N. Diagnostic difficulties with Plasmodium knowlesi infection in humans. Emerg Infect Dis. 2010;16:1033–4.

    PubMed Central 

    Google Scholar 

  • Sato S. Plasmodium—a brief introduction to the parasites causing human malaria and their basic biology. J Physiol Anthropol. 2021;40:1.

    PubMed Central 

    Google Scholar 

  • Davidson G, Speldewinde P, Manin BO, Cook A, Weinstein P, Chua TH. Forest restoration and the zoonotic vector Anopheles balabacensis in Sabah, Malaysia. EcoHealth. 2024;21:21–37.

    Google Scholar 

  • Froeschl G, Beissner M, Huber K, Bretzel G, Hoelscher M, Rothe C. Plasmodium knowlesi infection in a returning German traveller from Thailand: a case report on an emerging malaria pathogen in a popular low-risk travel destination. BMC Infect Dis. 2018;18:148.

    PubMed Central 

    Google Scholar 

  • Shearer FM, Huang Z, Weiss DJ, Wiebe A, Gibson HS, Battle KE, et al. Estimating geographical variation in the risk of zoonotic Plasmodium knowlesi infection in countries eliminating malaria. PLoS Negl Trop Dis. 2016;10: e0004915.

    PubMed Central 

    Google Scholar 

Continue Reading

  • Paolo Sorrentino's 'La Grazia' to open Venice Film Festival – Reuters

    1. Paolo Sorrentino’s ‘La Grazia’ to open Venice Film Festival  Reuters
    2. Paolo Sorrentino’ La Grazia is the Opening Film of the Biennale Cinema 2025  La Biennale di Venezia
    3. Paolo Sorrentino’s ‘La Grazia’ Toplining ‘The Great Beauty’ Star Toni Servillo Set as Venice Film Festival Opener  Variety
    4. Paolo Sorrentino’s ‘La Grazia’ To Open Venice Film Festival  Deadline
    5. Sorrentino’s new film La Grazia to open Venice Film Festival  ANSA

    Continue Reading

  • Bionomics of Anopheles gambiae complex (Diptera: Culicidae) and malaria transmission pattern in a pre-elimination area in South–Western Senegal | Malaria Journal

    Bionomics of Anopheles gambiae complex (Diptera: Culicidae) and malaria transmission pattern in a pre-elimination area in South–Western Senegal | Malaria Journal

  • WHO. Guidelines for malaria vector control. Geneva: World Health Organization; 2019. p. 1–161.

    Google Scholar 

  • WHO. World Malaria Report 2022. Geneva: World Health Organization; 2022. p. 1–372.

    Google Scholar 

  • NMCP. Annual Epidemiological Newsletter 2021 on Malaria in Senegal. Dakar: Ministry of Health and Social Action; 2022.

    Google Scholar 

  • NMCP. Annual Epidemiological Newsletter 2022 on Malaria in Senegal. Dakar: Ministry of Health and Social Action; 2023.

    Google Scholar 

  • Sene S, Ozer P. Rainfall trends and relationship floods and rainy events in Senegal. Geogr Soc Newsl Liege. 2002;42:27–33.

    Google Scholar 

  • Diouf I, Deme A, Rodriguez Fonseca B, Cisse M, Ndione JA, Gaye AT. Determination of malaria parameters in Senegal using meteorological station data and reanalysis. Guayaquil: XXVIIIe Colloquium of the International Association of Climatology; 2015.

    Google Scholar 

  • Faye O, Gaye O, Fontenille D, Sy N, Konate L, Hébrard G, et al. Comparison of malaria transmission in two epidemiological facies in Senegal: the Sahelian coastal zone and the southern Sudanian zone. Dakar Med. 1995;40:201–7 (in French).

    CAS 

    Google Scholar 

  • Faye O, Konaté L, Diop A. Entomological profile of malaria in Senegal. Dakar: Ministry of Health Medical Prevention; 2011.

    Google Scholar 

  • Fleury L, Pison G, Delaunay V. Senegal-Mlomp health and population observatory (1985–ongoing). Arch Natl Data Senegal. 2019;1–12.

  • PDC. Diagnostic report for the Commune of Mlomp. Report. 2018. http://www.pouyelayese.com/ardzig/media/attachments/2020/11/23/rapport-final_pdc_mlomp-ouss.pdf.

  • CDO. Departmental land use and development plan of Oussouye. Report. 2016;1–90.

  • Diagne N, Fontenille D, Konate L, Faye O, Lamizana MT, Legros F, et al. The Anopheline of Senegal: an annotated and illustrated list. Bull Soc Path Exot. 1994;87:267–77 (in French).

    CAS 

    Google Scholar 

  • Detinova TS. Determination of the physiological age of female Anopheles from the changes of the tracheal system of the ovaries. Med Parazitol (Mosk). 1945;14:45–9 (in Russian).

    CAS 

    Google Scholar 

  • Beier JC, Perkins PV, Wirtz RA, Koros J, Diggs D, Gargan TP, et al. Bloodmeal identification by direct enzyme-linked immunosorbent assay (ELISA), tested on Anopheles (Diptera: Culicidae) in Kenya. J Med Entomol. 1988;25:9–16.

    CAS 

    Google Scholar 

  • Wirtz RA, Zavala F, Charoenvit Y, Campbell GH, Burkot TR, Schneider I, et al. Comparative testing of monoclonal antibodies against Plasmodium falciparum sporozoites for ELISA development. Bull World Health Organ. 1987;65:39–45.

    CAS 

    Google Scholar 

  • Collins FH, Mendez MA, Rasmussen MO, Mehaffey PC, Besansky NJ, Finnerty V. A ribosomal RNA gene probe differentiates member species of the Anopheles gambiae complex. Am J Trop Med Hyg. 1987;37:37–41.

    CAS 

    Google Scholar 

  • Wilkins EE, Howell PI, Benedict MQ. IMP PCR primers detect single nucleotide polymorphisms for Anopheles gambiae species identification, Mopti and Savanna rDNA types, and resistance to dieldrin in Anopheles arabiensis. Malar J. 2006;5:125.

    Google Scholar 

  • Fontenille D, Faye O, Konate L, Sy N, Collins FH. Comparison of PCR and cytogenetic techniques to identify members of the Anopheles gambiae complex in Senegal. Ann Parasitol Hum Comp. 1993;68:239–40 (in French).

    CAS 

    Google Scholar 

  • Ndiath MO, Brengues C, Konate L, Sokhna C, Boudin C, Trape JF, et al. Dynamics of transmission of Plasmodium falciparum by Anopheles arabiensis and the molecular forms M and S of Anopheles gambiae in Dielmo, Senegal. Malar J. 2008;7:136.

    Google Scholar 

  • Caputo B, Tondossoma N, Virgillito C, Pichler V, Serini P, Calzetta M, et al. Is Côte d’Ivoire a new high hybridization zone for the two major malaria vectors, Anopheles coluzzii and An. gambiae (Diptera, Culicidae)? Infect Genet Evol. 2022;98:105215.

    Google Scholar 

  • Nwakanma DC, Neafsey DE, Jawara M, Adiamoh M, Lund E, Rodrigues A, et al. Breakdown in the process of incipient speciation in Anopheles gambiae. Genetics. 2013;193:1221–31.

    Google Scholar 

  • Oliveira E, Salgueiro P, Palsson K, Vicente JL, Arez AP, Jaenson TG, et al. High levels of hybridization between molecular forms of Anopheles gambiae from Guinea Bissau. J Med Entomol. 2008;45:1057–63.

    CAS 

    Google Scholar 

  • Sougoufara S, Sokhna C, Diagne N, Doucouré S, Sembène PM, Harry M. The implementation of long-lasting insecticidal bed nets has differential effects on the genetic structure of the African malaria vectors in the Anopheles gambiae complex in Dielmo, Senegal. Malar J. 2017;16:337.

    Google Scholar 

  • Niang EHA, Konaté L, Diallo M, Faye O, Dia I. Reproductive isolation among sympatric molecular forms of An. gambiae from inland areas of South-Eastern Senegal. PLoS ONE. 2014;9: e104622.

    Google Scholar 

  • Caputo B, Nwakanma D, Jawara M, Adiamoh M, Dia I, Konate L, et al. Anopheles gambiae complex along The Gambia river, with particular reference to the molecular forms of An. gambiae s.s. Malar J. 2008;7:17.

    Google Scholar 

  • Della Torre A, Tu Z, Petrarca V. On the distribution and genetic differentiation of Anopheles gambiae s.s. molecular forms. Insect Biochem Mol Biol. 2005;35:755–69.

    CAS 

    Google Scholar 

  • Niang A, Epopa PS, Sawadogo SP, Maïga H, Konaté L, Faye O, et al. Does extreme asymmetric dominance promote hybridization between Anopheles coluzzii and Anopheles gambiae s.s. in seasonal malaria mosquito communities of West Africa. Parasit Vectors. 2015;8:586.

    Google Scholar 

  • Faye O, Konate L, Mouchet J, Fontenille D, Sy N, Hebrard G, et al. Indoor resting by outdoor biting females of Anopheles gambiae complex (Diptera: Culicidae) in the Sahel of Northern Senegal. J Med Entomol. 1997;34:285–9.

    CAS 

    Google Scholar 

  • Faye O. Malaria in Senegal. Ecology of the parasitosis and perspectives for control. Thesis, Nat Univ Cheikh Anta Diop Dakar. 1994.

  • Niang EHA, Touré A, Ngom EHM, Konaté L, Faye O, Diallo M, et al. Malaria transmission pattern in an area selected for clinical trials in the Sudanian area of Senegal (West Africa). J Trop Med. 2013;2013: 907375.

    Google Scholar 

  • Dia I, Diop T, Rakotoarivony I, Kengne P, Fontenille D. Bionomics of Anopheles gambiae Giles, An. arabiensis Patton, An. funestus Giles and An. nili (Theobald) (Diptera: Culicidae) and Transmission of Plasmodium falciparum in a Sudano-Guinean Zone (Ngari, Senegal). J Med Entomol. 2003;40:279–83.

    Google Scholar 

  • Rajaonarivelo V, Le Goff G, Cot M, Brutus L. Anopheline and malaria transmission in Ambohimena, a village on the western margin of the Malagasy Highlands. Parasite. 2004;11:75–82.

    CAS 

    Google Scholar 

  • Assouho KF, Adja AM, Guindo-Coulibaly N, Tia E, Kouadio AMN, Zoh DD, et al. Vectorial transmission of malaria in major districts of Côte d’Ivoire. J Med Entomol. 2020;57:908–14.

    Google Scholar 

  • Faye O. Contribution to the study of Anophelinae (Diptera, Culicidae) and malaria transmission in the area of the Bignona anti-salt dam (Ziguinchor, Senegal). Thesis. Univ Cheikh Anta Diop Dakar. 1987.

  • Robert V, Dieng H, Lochouarn L, Traoré SF, Trape J-F, Simondon F, et al. Malaria transmission in the Niakhar area, Senegal. Trop Med Int Health. 1998;3:667–77.

    CAS 

    Google Scholar 

  • Faye O, Gaye O, Hervé JP, Diack PA, Diallo S. Malaria in the Sahelian zone of Senegal. Ann Soc Belg Med Trop. 1993;73:21–30.

    CAS 

    Google Scholar 

  • Konate L, Diagne N, Brahimi K, Faye O, Legros F, Rogier C, et al. Vector biology and transmission of Plasmodium falciparum, P. malariae et P. ovale in a West African savannah village (Dielmo, Senegal). Parasite. 1994;1:325–33.

    CAS 

    Google Scholar 

  • Faye O, Fontenille D, Gaye O, Sy N, Molez JF, Diallo S, et al. Malaria and rice growing in the Senegal River delta (Senegal). Ann Soc Belg Med Trop. 1995;75:179–89.

    CAS 

    Google Scholar 

  • Yadouleton A, Aikpon R, Houndeton G, Aboubacar S, Ursins F, Tchibçozo C, et al. Preliminary entomological data for the implementation of large-scale indoor residual spraying in the commune of Corpargo in Northeastern Benin. Int J Biol Chem Sci. 2018;12:1993–2003.

    CAS 

    Google Scholar 

  • Carrara GC, Petrarca V, Niang M, Coluzzi M. Anopheles pharoensis and transmission of Plasmodium falciparum in the Senegal River delta, West Africa. Med Vet Entomol. 1990;4:421–4.

    CAS 

    Google Scholar 

  • Manga L, Toto JC, Carnevale P. Vectors and malaria transmission around the new Yaounde-Nsimalen international airport. Bull Liaison Doc-OCEAC. 1992;102:48–55.

    Google Scholar 

  • Diop A, Molez JF, Konaté L, Fontenille D, Gaye O, Diouf M, et al. Role of Anopheles melas Theobald (1903) in malaria transmission in the Saloum mangrove (Senegal). Parasite. 2002;9:239–46.

    CAS 

    Google Scholar 

  • Jawara M, Pinder M, Drakeley CJ, Nwakanma DC, Jallow E, Bogh C, et al. Dry season ecology of Anopheles gambiae complex mosquitoes in The Gambia. Malar J. 2008;7:156.

    Google Scholar 

  • Akogbeto M. Entomological study of coastal lagoon malaria transmission: the case of a village built on a brackish-water lake. Ann Soc Belg Med Trop. 1995;75:219–27.

    CAS 

    Google Scholar 

  • Trape JF, Pison G, Preziosi MP, Enel C, Du Loû AD, Delaunay V, et al. Impact of chloroquine resistance on malaria mortality. C R Acad Sci. 1998;III(321):689–97.

    Google Scholar 

  • Agnamey P, Brasseur P, Eldin de Pecoulas P, Vaillant M, Olliaro P. Plasmodium falciparum in vitro susceptibility to antimalarial drugs in Casamance (South-western Senegal) during the first 5 years of routine use of artesunate-amodiaquine. Antimicrob Agents Chemother. 2006;50:1531–4.

    CAS 

    Google Scholar 

  • Continue Reading

  • Gold poised for weekly gain on dollar weakness, safe-haven demand – Reuters

    1. Gold poised for weekly gain on dollar weakness, safe-haven demand  Reuters
    2. Gold price remains on track to register gains for the first time in three weeks  FXStreet
    3. Gold (XAUUSD) Price Forecast: Gold Market Eyes Breakout on Dollar Dip, Tariff Fears  FXEmpire
    4. Gold heads for weekly gain as US tax-cut bill stokes fiscal worries  Dunya News
    5. Gold falls as strong US payrolls data douses rate cut hopes  Business Recorder

    Continue Reading

  • Long neglected on dusty shelves in Corsican bars, Cap Corse is now looking to make an international comeback

    Long neglected on dusty shelves in Corsican bars, Cap Corse is now looking to make an international comeback

    After a period of decline, the island’s once-iconic apéritif is experiencing a notable resurgence by looking to the past.

    In Jacques Deray’s 1974 film Borsalino & Cie, Alain Delon, the epitome of French cool in his hat and double-breasted suit, takes revenge on his enemies in glamorous locales, where dapper card sharks enjoy generous pours of ruby-red Cap Corse. After decades confined to the dusty shelves of old-fashioned bars on Napoleon’s native island, this Corsican liqueur is spritzing its way back to prominence.

    Cap Corse, which comes in white and red variants, is a wine-based drink infused with tree bark, citrus fruit and herbs. LN Mattei, the company that distils it, was founded in the early 1870s. The tipple reached the peak of its popularity during the early 20th century. In the decades after the Second World War, however, it steadily lost its lustre, becoming a drink that mostly appealed to older Corsicans who remembered its glory days. 

    In 2016, Corsica’s Groupe Boisson Corse acquired LN Mattei and kick-started a new era of expansion. It has gone from producing 80,000 bottles, sold almost exclusively in Corsica, to making 400,000 bottles today, 15 per cent of which are exported.

    “Over time, Cap Corse had evolved to emphasise the bitterness of cinchona bark,” says Patrice Gontier Ackermann, LN Mattei’s general manager. “To appeal to a broader audience, we revived the original recipe of our founder, Louis Napoléon Mattei, which balances sweetness and bitterness.” This change was informed by the tastes of the current golden age of mixology but also by the rise of the Aperol spritz, which, in less than a decade, has gone from a Venetian aperitivo to a global phenomenon. (Aperol is now the most valuable brand in the Campari group’s European portfolio.) “We recognised that offering a quirky Corsican alternative had significant potential,” says Gontier Ackermann.

    The “Capo Spritz” is now served in bars across the island in large LN Mattei-branded glasses that will look familiar to Aperol fans. Expect to see these gracing a Mediterranean bar this summer. With just 350,000 or so permanent residents in Corsica and near-universal brand recognition on the island, LN Mattei needs to lean hard on exports for future growth. By 2030 it plans to double production to 800,000 bottles, with half sold outside Corsica.

    Cap Corse’s resurgence mirrors the island’s rise as a destination, increasingly attracting tourists from beyond France. The number of foreign visitors was up by 6 per cent last year, hitting an all-time high. LN Mattei’s shop in Bastia’s Place Saint Nicolas is an especially strong asset. Established by the company’s founder in 1872, it’s an officially recognised historical landmark. The shop, known for its dark-red open cabinets and exposed stone walls, stocks a wide array of Corsican products alongside the booze.

    An alcohol-free version of the apéritif is currently in the works. “That could become an important market for us in the years to come,” says Gontier Ackermann. After long being overlooked, Cap Corse is seeking to shake things up. “If we don’t keep moving, we could disappear.”

    Continue Reading

  • Shehbaz Sharif meets Iranian president on sidelines of ECO summit

    Shehbaz Sharif meets Iranian president on sidelines of ECO summit

    Prime Minister Shehbaz Sharif met with Iranian President Dr Masoud Pezeshkian on Friday on the sidelines of the 17th Economic Cooperation Organization (ECO) Summit held in Khankendi, Azerbaijan.

    During the meeting, the two leaders reviewed ongoing bilateral cooperation across all sectors and expressed satisfaction over the progress made on decisions taken during their previous meeting aimed at further strengthening Pakistan-Iran relations.

    The two leaders also discussed the evolving regional situation in light of Israel’s unjustified aggression against Iran. PM Shehbaz appreciated President Pezeshkian’s leadership and commended Iran’s decision to exercise restraint and pursue a ceasefire during the recent crisis.

    Reiterating Pakistan’s unwavering solidarity with the people and government of Iran, the premier reaffirmed Pakistan’s firm commitment to working together with Iran through dialogue and diplomacy for peace in the region.

    Read: PM to attend 17th ECO summit in Baku

    President Pezeshkian appreciated Pakistan’s strong diplomatic support for Iran at international forums during the recent crisis and thanked Pakistan for its key role in de-escalation efforts.

    PM Shehbaz also conveyed a message of congratulations and best wishes for Iran’s Supreme Leader Ayatollah Ali Khamenei.

    17th ECO Summit

    PM Shehbaz reached Azerbaijan on Thursday for a two-day official visit to participate in the 17th Summit of the ECO.

    The summit, was held in Khankendi from July 3 to 4, under the theme “New ECO Vision for a Sustainable and Climate Resilient Future”, bringing together heads of state and government from member countries to discuss pressing regional issues, including connectivity, trade and climate change.

    The premier was accompanied by Deputy Prime Minister and Foreign Minister Ishaq Dar, Federal Information Minister Attaullah Tarar and Special Assistant to the Prime Minister Tariq Fatemi.

    Upon his arrival at Fuzuli International Airport, the prime minister was received by Azerbaijan’s Minister of Culture Adil Karimli, Azerbaijan’s Ambassador to Pakistan Khazar Farhadov, Pakistan’s Ambassador to Azerbaijan Qasim Moinuddin and other senior diplomatic and government officials.

    The Foreign Office had stated that the prime minister would address the summit outlining Pakistan’s perspective on key regional and global issues. He was expected to reaffirm Pakistan’s commitment to the ECO Vision 2025 and advocate for deeper intra-regional trade, enhanced transport connectivity, greater energy cooperation and sustainable development.

    Continue Reading

  • NIS2 revamps Ireland’s cybersecurity landscape: Old regulators, new powers

    The NIS2 Directive has significantly reshaped the cybersecurity landscape across the EU. Since the implementation deadline in October 2024, EU Member States have been working to incorporate new standards into their national laws, fostering a dynamic and rapidly evolving regulatory environment. Recently, Ireland’s National Cyber Security Centre (NCSC) published the draft NIS2 Risk Management Measures (RMM) Guidance, which outlines the minimum requirements for essential and important entities.  Ireland has also joined the Cyber Fundamentals Framework (CyFun), originally developed in Belgium, as a scheme co-owner. The CyFun framework offers a structured, risk-based methodology for essential and important entities, assisting them in organising and demonstrating their NIS2 security measures.

    While Ireland has not yet implemented the NIS2 Directive, the legislative process is now at an advanced stage, with the NCSC expecting the National Cyber Security Bill to be transposed into law by the end of the year. As implementation is steadily advancing, it is important to consider how it will impact businesses operating in and through Ireland.

    NIS2’s overall goal is to achieve a high common level of cybersecurity across the EU. Part of this approach requires Member States to establish one or more Competent Authorities responsible for cybersecurity and enforcement. To ensure smooth cross border compliance, a Single Point of Contact on cybersecurity responsible for liaising with other Member States is also required.

     

    Ireland’s NIS2 Strategy: NCSC as Central Liaison with Sectoral Oversight

    Interestingly, Ireland’s proposed implementing legislation takes a different approach to what is outlined in the NIS2 Directive. The NCSC will serve as the Single Point of Contact and act as the Lead Competent Authority, providing advice, guidance, and support to a range of Competent Authorities, each of which will oversee enforcement within their respective sectors. This approach contrasts with Belgium and France for example, where the Centre for Cyber Security Belgium (CCB) and the French Cyber Security Agency (ANSSI) serve as the central authority for all cybersecurity matters. Ireland’s NCSC will act as an overseer. An explanatory note in the Heads of Bill acknowledges that this role does not exist within the NIS2 Directive and “was taken as a policy decision after engagement with the other competent authorities in agreement with the NCSC”.

     

    Regulator Overview

    Article 8 of the NIS2 Directive mandates the designation of Competent Authorities, providing for robust oversight and enforcement. Article 27 requires businesses within the scope of the NIS2 Directive to register with these authorities, ensuring compliance and accountability. The full list of National Competent Authorities along with their designated sectors, as set out in head 17 of the draft Bill is as follows:

     

    NIS2 Regulatory and Oversight Bodies in Ireland
    Sectors
    Commission for the Regulation of Utilities (CRU)
    • Energy
    • Drinking Water
    • Wastewater
    Commission for Communications Regulation (ComReg)
    • Digital Infrastructure
    • ICT Service Management
    • Digital Providers
    • Space
    Central Bank of Ireland (CBI)
    Irish Aviation Authority (IAA)
    Commission for Rail Regulation (CRR)
    The Minister for Transport
    National Transport Authority (NTA)
    Agencies under the remit of the Minister for Health
    National Cyber Security Centre (NCSC)
    • All other sectors set out in Schedule I and II
     
    Main Establishment & Considerations for Digital Service Providers

    The main establishment provision within the NIS2 Directive aims to reduce jurisdictional conflicts and regulatory overlap in the digital services space by creating a ‘one-stop-shop’ for incident reporting obligations. The main benefit derived from these provisions is that entities can report incidents to a single Member State rather than multiple jurisdictions, streamlining compliance and reducing the reporting burden for entities in the midst of large-scale cyber incidents.

    Digital Service Providers looking to avail of the main establishment concept within the NIS2 Directive should note that the Commission for Communications Regulation (ComReg) will be responsible for this sector in Ireland. ComReg has been noticeably proactive in issuing guidance and developing resources in relation to the NIS2 Directive, indicating that it is preparing to play a significant role in enforcement – this will be welcomed by many businesses, given Ireland’s considerable importance as the European or EMEA headquarters for many of the world’s major tech companies.

    Established in 2002, ComReg is a mature, well-resourced statutory body which is responsible for regulating electronic communications and the postal sector in Ireland. ComReg has been instrumental in ensuring compliance throughout the most critical sectors in the Irish economy. Until recently, ComReg was under the leadership of Helen Dixon, who formerly led the Data Protection Commission and was instrumental in its transformation from a small, regionally based office to one of the world’s most influential data protection regulators.

     

    Conclusion

    As Ireland moves closer to full implementation of the NIS2 Directive, we are embracing a distinctive regulatory model that balances central coordination with sector-specific oversight. The NCSC’s role as both the Single Point of Contact and Lead Competent Authority reflects a pragmatic approach tailored to Ireland’s regulatory landscape. With the publication of the draft RMM Guidance and Ireland’s co-ownership of the CyFun framework, the groundwork is being laid for a more structured and resilient cybersecurity regime.

    Proactive engagement with sectoral regulators, especially ComReg for digital services, will be essential to ensure compliance and to leverage the benefits of streamlined reporting and guidance. Digital services providers can expect a well-resourced, tough but fair regulator capable of engaging with both multinational and national businesses. Some digital services providers may be classified as highly critical and subject to stricter enforcement, including proactive measures such as audits. The specifics of enforcement will become clear once the NIS2 Directive is transposed.

    As the National Cyber Security Bill nears enactment, organisations should prepare for a more robust and coordinated cybersecurity environment that aligns with the EU’s broader vision of digital resilience and cross-border cooperation. For further information, please do not hesitate to get in touch with one of the authors or your usual DLA Piper contact.

    Continue Reading