Skip to main content
Advertisement
  • Loading metrics

The endosymbiont Wolbachia rebounds following antibiotic treatment

  • Emma L. Gunderson,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Project administration, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • Ian Vogel,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Project administration, Writing – original draft, Writing – review & editing

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • Laura Chappell,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Project administration, Visualization, Writing – review & editing

    Affiliation Dept. of Molecular, Cell and Developmental Biology; University of California, Santa Cruz; Santa Cruz, California, United States of America

  • Christina A. Bulman,

    Roles Formal analysis, Investigation, Methodology, Writing – review & editing

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • K. C. Lim,

    Roles Investigation, Methodology

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • Mona Luo,

    Roles Investigation, Methodology

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • Jeffrey D. Whitman,

    Roles Investigation, Methodology, Writing – review & editing

    Affiliation Dept. of Laboratory Medicine; University of California, San Francisco; San Francisco, California, United States of America

  • Chris Franklin,

    Roles Investigation, Methodology

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • Young-Jun Choi,

    Roles Investigation, Methodology, Writing – review & editing

    Affiliation Division of Infectious Diseases; Washington University School of Medicine, St. Louis; St. Louis, Missouri, United States of America

  • Emilie Lefoulon,

    Roles Formal analysis, Investigation, Methodology

    Affiliation Molecular Parasitology Division; New England BioLabs; Ipswich, Massachusetts, United States of America

  • Travis Clark,

    Roles Formal analysis, Investigation, Methodology, Writing – review & editing

    Affiliation Veterinary Pathobiology; University of Missouri-Columbia; Columbia, Missouri, United States of America

  • Brenda Beerntsen,

    Roles Formal analysis, Investigation, Methodology, Resources, Writing – review & editing

    Affiliation Veterinary Pathobiology; University of Missouri-Columbia; Columbia, Missouri, United States of America

  • Barton Slatko,

    Roles Conceptualization, Methodology, Supervision, Writing – original draft, Writing – review & editing

    Affiliation Molecular Parasitology Division; New England BioLabs; Ipswich, Massachusetts, United States of America

  • Makedonka Mitreva,

    Roles Conceptualization, Formal analysis, Funding acquisition, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing

    Affiliation Division of Infectious Diseases; Washington University School of Medicine, St. Louis; St. Louis, Missouri, United States of America

  • William Sullivan,

    Roles Conceptualization, Formal analysis, Funding acquisition, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing

    Affiliation Dept. of Molecular, Cell and Developmental Biology; University of California, Santa Cruz; Santa Cruz, California, United States of America

  •  [ ... ],
  • Judy A. Sakanari

    Roles Conceptualization, Formal analysis, Funding acquisition, Methodology, Project administration, Supervision, Visualization, Writing – original draft, Writing – review & editing

    judy.sakanari@ucsf.edu

    Affiliation Dept. of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, California, United States of America

  • [ view all ]
  • [ view less ]

Abstract

Antibiotic treatment has emerged as a promising strategy to sterilize and kill filarial nematodes due to their dependence on their endosymbiotic bacteria, Wolbachia. Several studies have shown that novel and FDA-approved antibiotics are efficacious at depleting the filarial nematodes of their endosymbiont, thus reducing female fecundity. However, it remains unclear if antibiotics can permanently deplete Wolbachia and cause sterility for the lifespan of the adult worms. Concerns about resistance arising from mass drug administration necessitate a careful exploration of potential Wolbachia recrudescence. In the present study, we investigated the long-term effects of the FDA-approved antibiotic, rifampicin, in the Brugia pahangi jird model of infection. Initially, rifampicin treatment depleted Wolbachia in adult worms and simultaneously impaired female worm fecundity. However, during an 8-month washout period, Wolbachia titers rebounded and embryogenesis returned to normal. Genome sequence analyses of Wolbachia revealed that despite the population bottleneck and recovery, no genetic changes occurred that could account for the rebound. Clusters of densely packed Wolbachia within the worm’s ovarian tissues were observed by confocal microscopy and remained in worms treated with rifampicin, suggesting that they may serve as privileged sites that allow Wolbachia to persist in worms while treated with antibiotic. To our knowledge, these clusters have not been previously described and may be the source of the Wolbachia rebound.

Author summary

Onchocerciasis (river blindness) and lymphatic filariasis (elephantiasis) are two neglected tropical diseases caused by filarial nematodes, which harbor the endosymbiotic bacteria, Wolbachia. Major efforts to discover new drugs to treat these diseases have led to the discovery of novel compounds including those that target Wolbachia. We investigated the long-term effects of rifampicin on the filarial nematode, Brugia pahangi, and its endosymbiont, Wolbachia, in an in vivo rodent model of infection. Initially, Wolbachia titers were significantly reduced by 95% and female fecundity was impaired shortly after treatment. 8 months later however, Wolbachia rebounded and embryogenesis returned to normal. Sequence analysis of the Wolbachia genome revealed that despite the population bottleneck and recovery, no genetic changes occurred that could account for the rebound. Clusters of Wolbachia were observed within the ovaries of female worms throughout the entire 8-month study. These clusters may sequester Wolbachia and allow the bacteria to persist during antibiotic treatment, thereby enabling them to repopulate ovarian tissues and ensuring their vertical transmission to future generations of microfilariae.

Introduction

Onchocerciasis, commonly known as river blindness, and lymphatic filariasis (LF), commonly known as elephantiasis, are neglected tropical diseases caused by filarial worms that together affect an estimated 86 million people worldwide [1]. Approximately 1.2 million people are visually impaired due to river blindness, while 12 million people with LF have complications due to elephantiasis [1]. River blindness is caused by the release of thousands of microfilariae (mf) from adult Onchocerca volvulus females residing in subcutaneous tissues. Mf migrate through the skin causing severe itching and skin depigmentation and also migrate to the ocular region where they induce an inflammatory response that can lead to blindness [2]. LF is caused by Wuchereria bancrofti, Brugia malayi and B. timori worms that reside in the lymphatic tissues where they cause tissue damage. While many infections are asymptomatic, individuals that develop the disfiguring disease often experience pain and severe lymphedema typically in the arms, legs, breasts and genitalia [2]. This can result in stigma associated with elephantiasis and extreme economic loss for individuals suffering from this disease [3]. The years lived with disability (YLDs) for LF and onchocerciasis are estimated to be 1.4 million and 1.3 million, respectively [1].

Current treatments primarily target the mf and not the adult worms, which are capable of surviving in their human host for 10–14 years for O. volvulus and 6–8 years for Brugia spp. [2,49]. For this reason, international control programs require annual or biannual mass drug administration of drugs in order to reduce transmission rates. However, given the longevity and high fecundity of these worms and the current lack of drugs that kill the adult worms, it is unlikely that the WHO goal of eliminating LF and onchocerciasis by 2030 will be met when microfilaricidal drugs are used alone [1015]. The inability to reduce transmission rates with microfilaricides is compounded by the fact that ivermectin (IVM) cannot be distributed in areas co-endemic for another filarial nematode, Loa loa, due to the risk of severe adverse events, especially toxic encephalopathy when individuals are co-infected with high loads of L. loa mf [16,17].

Onchocerca, Wuchereria and Brugia spp., like many other species of filarial nematodes, harbor an intracellular endosymbiont, Wolbachia, which is important for female worm fecundity and survival [1822]. Loa loa, however, lacks this bacterium, and efforts are underway to develop anti-Wolbachia drugs to eliminate this bacterium, thereby resulting in death of adult worms. In clinical trials conducted on patients with onchocerciasis and lymphatic filariasis, doxycycline was shown to deplete Wolbachia and eventually eliminate the adult worms after about 1–2 years [23,24]. Doxycycline however requires lengthy dosing regimens (100–200 mg daily for 4–6 weeks) and is therefore not practical for mass drug distribution. In addition, doxycycline is contraindicated in children 8 years and younger and because it is in pregnancy category D, should not be given to pregnant women [2].

Several studies have recently shown that short courses of 7- and 14-days of anti-Wolbachia compounds hold promise as excellent drugs to treat onchocerciasis and LF [2529]. Studies by Hübner et al. however, showed that suboptimal treatment regimens of doxycycline in the Litomosoides sigmodontis infection model did not lead to a sustained reduction in Wolbachia loads in worms 14–18 weeks post-treatment and that longer term studies were needed to assess permanent sterilization of female adult worms [27,29]. Although West African cattle infected with Onchocerca ochengi are excellent hosts for long-term studies to evaluate the efficacy of antibiotics for the treatment of human onchocerciasis [3036], the purpose of our study was to investigate the long-term effects of rifampicin in a rodent model of infection. In the present study, we investigated the use of jirds infected with Brugia pahangi to assess the effects of rifampicin on Wolbachia and worm survival in an 8-month time course study in which Wolbachia titers were determined using adult worms recovered from animals treated with a one-week dosing regimen of rifampicin.

Results

The endosymbiont Wolbachia rebound after 8 months following rifampicin treatment without genetic change

The research objective was to determine the long-term effects of antibiotic treatment on filarial worms and their endosymbiont, Wolbachia. We hypothesized that rifampicin would deplete worms of Wolbachia which would eventually lead to adult worm death in jirds infected with Brugia pahangi. Wolbachia titers in adult male and female worms were determined by qPCR at 1 week, 6 weeks, 17 weeks and 8 months post-first dose following the protocol by McGarry et al [37]. The relative abundance of single copy genes encoding the Wolbachia surface protein (wsp) were normalized to that of Brugia glutathione-S-transferase gene (gst) [28,29,3840]. At the 1-week timepoint, Wolbachia titers were significantly reduced by 95.2% in female worms (Fig 1A). By 6 weeks however, the reduction in Wolbachia titers was reduced by 81.3% compared to those of control worms and by 17 weeks, titers were reduced by 77%. At 8 months, Wolbachia titers returned to levels similar to those of control worms, i.e. there was a 0% reduction in Wolbachia titers (Fig 1A). Wolbachia titers from male worms followed a similar trend of rebound (Fig 1B).

thumbnail
Fig 1. Wolbachia titers return to control levels 8 months after rifampicin treatment.

Female and male Brugia pahangi from jirds treated with rifampicin were analyzed by qPCR to determine Wolbachia titers at each timepoint. Mean percent reductions of Wolbachia wsp/gst ratios from female (A) and male (B) adult worms are shown at 1-week, 6-weeks, 17-weeks and 8-months post-first dose. Data shown are medians and the boxes are the 25th and 75th percentiles with ***P<0.001 and *P<0.05. n = 2–9 jirds per treatment group per timepoint. Additional information is shown in S1 Table.

https://doi.org/10.1371/journal.ppat.1008623.g001

Although there was a significant effect of rifampicin on Wolbachia titers at early timepoints (S1 Table), rifampicin did not reduce the number of adult worms recovered at the time of necropsy at any of the timepoints, i.e. no macrofilaricidal effects were observed (S1 Fig).

To determine if the rebound in Wolbachia occurred as a result of genome changes (e.g. antibiotic resistance/tolerance), we sequenced Wolbachia genomes using hybridization probe-capture method from the treatment and vehicle groups at the 1-week and 8-month timepoints [41]. On average ~90,000 PacBio CCS reads were generated per sample, which amounts to ~150× coverage of the genome (S2 Table). A complete circularized reference genome (1,072,983 bp) was assembled using the 1-week vehicle group and sequence variants were identified in each group with respect to the reference. Variants occurring within the genomic regions likely representing nuclear Wolbachia transfers (nuwts [42]) were excluded from the analysis.

One single nucleotide variant (SNV) and five insertion/deletion variants (INDEL) were identified and their allele frequencies were estimated based on the number of reads that support each allele in each sample (S3 Table). The SNV (C-to-T substitution) occurred within the ORF of a short-chain dehydrogenase/reductase family (SDR) oxidoreductase and was predicted to be a synonymous variant and therefore a silent mutation. The allele frequency of this variant increased from 5.1% in the 1-week vehicle group to 35.5% in the 8-month rifampicin group (Fisher’s exact test P-value: 3.2×10−5), which was the only variant whose allele frequency displayed a statistically significant change. The INDEL variants invariably occurred within homopolymer regions (9–13 consecutive bases of A or T). Homopolymeric tracts are mutational hotspots because they are vulnerable to slippage errors during replication and transcription [43]. However, INDEL calling is error-prone around homopolymer runs (due to sequencing and PCR errors), and we cannot exclude the possibility that these INDELs are false-positive variants [44]. These data suggest that, despite the population bottleneck and recovery, the genetic change in Wolbachia after rifampicin treatment likely did not occur.

Female worm fecundity is significantly reduced shortly after treatment but returns to control levels by 17 weeks

Commensurate with the depletion of Wolbachia 1 week after the first dose of rifampicin, we observed a significant impact on embryogenesis 6 weeks post-antibiotic treatment. This was followed by a gradual rebound in Wolbachia and return to normal embryogenesis by 17 weeks.

The fecundity of female B. pahangi at each timepoint was assessed by counting the number of mf released after worms were removed from the animals and incubated in vitro for 18 hours. Worms from rifampicin treated jirds at the 1- and 6-week timepoint showed a significant reduction in the number of mf that were shed compared to worms from the vehicle group (43.4% reduction, P<0.05 and 86.3% reduction, P<0.0001, respectively) (Fig 2). Female fecundity returned to control levels after 17 weeks and remained at control levels for up to 8 months.

thumbnail
Fig 2. Rifampicin decreases mf shedding from female worms up to 6 weeks, followed by a return to normal by 17 weeks.

The number of mf shed overnight by adult female worms that were recovered 1 week, 6 weeks, 17 weeks and 8-months post-first dose is shown for each timepoint. Mf shed overnight at the 1- and 6-week timepoint were significantly reduced (*P<0.05 and ****P<0.0001, respectively). Data are shown as median ± 95% CI. n = 10–26 female worms from n = 2–9 jirds per treatment group per timepoint.

https://doi.org/10.1371/journal.ppat.1008623.g002

Embryograms of female worms were also analyzed to determine the effects of rifampicin on the developing stages of mf within the reproductive tract of female worms. Results showed that developmental stages from female worms recovered from rifampicin treated jirds at the 6-week timepoint exhibited a significant decrease in healthy embryos and an increase in degenerated embryos compared to those from both the control group at the 6-week timepoint, and the rifampicin group at the 1-week timepoint, where little disruption was observed (Fig 3 and S4 Table). The decrease in fecundity and disruption of embryogenesis however were not observed at later timepoints suggesting that embryonic development of mf returned to control levels following the rebound of Wolbachia.

thumbnail
Fig 3. Rifampicin treatment leads to impaired embryogenesis by the 6-week timepoint but normal developmental stages return by 17 weeks.

Embryonic stages found within the ovaries and uteri from female worms from the vehicle and treated groups were counted 1 week, 6 weeks, 17 weeks and 8 months post-first dose. There was a significant decrease in the frequency of healthy embryos across all developmental stages of embryogenesis at the 6-week timepoint (****P<0.0001). Percentages of degenerated embryos (gray) were also determined for each timepoint. Data are presented as mean ± SEM. n = 6–9 female worms from n = 2–9 jirds per treatment group per timepoint.

https://doi.org/10.1371/journal.ppat.1008623.g003

Cellular analysis reveals the rebound is derived from clusters of Wolbachia

As an independent method of analyzing Wolbachia titer, we performed fluorescence confocal analysis to image Wolbachia and host cell nuclei as previously described by Landmann et al., Serbus et al. and Foray et al. [4549].

Fluorescence imaging of the distal tip region of the ovaries revealed that Wolbachia were nearly depleted from germline tissues at the 1-week timepoint, but they began to increase in number at later timepoints (Fig 4A). Interestingly, large densely packed “clusters” of Wolbachia (Fig 4B) were observed in worms recovered from both the vehicle and treated groups at each timepoint. When Wolbachia were quantified in each of the clusters from ovaries (Fig 4C), results showed there were no significant differences between the treated and control groups with respect to the Wolbachia density (puncta per area of cluster, Fig 4D). However, Wolbachia in the peripheral areas around the clusters were significantly reduced (P = 0.05) in the worms from the rifampicin group at 6-weeks but not at the later timepoints (Fig 4D).

thumbnail
Fig 4. Rebound of Wolbachia at later timepoints may be driven by clusters.

Ovaries were removed from individual female worms, fixed and stained for host nuclei (magenta) and Wolbachia (red). (A) Wolbachia are depleted at 1- and 6-weeks post-treatment but begin to rebound by 17 weeks. (B) Clusters of Wolbachia are seen in ovaries from vehicle and rifampicin worms; actin (green) stained with phalloidin; Wolbachia (red) stained with propidium iodide and host nuclei (magenta) stained with DAPI. (C) Wolbachia were quantified by counting number of puncta within the clusters (left) and in the periphery of the clusters (right). (D) Clusters were analyzed from worms collected at 6 weeks (n = 36–46), 17 weeks (n = 6–8) and 8 months (n = 34–52). Vehicle control worms in yellow and worms from rifampicin treated groups in red. Peripheral Wolbachia were reduced in females recovered from rifampicin treated jirds at the 6-week timepoint only (P = 0.05). Data are presented as median ± 95% CI.

https://doi.org/10.1371/journal.ppat.1008623.g004

The presence of clusters with densely packed Wolbachia in ovaries from both rifampicin treated and control worms suggests that despite clearance of bacteria from the areas outside the clusters, the Wolbachia nevertheless persisted within the clusters and may be the source of the rebound. Analyses were not conducted on male worms as the focus of the work was on female fecundity and embryogenesis. We postulate that similar clusters will be associated with male worms, possibly in the hypodermal areas and, as with females, are likely responsible for the rebound.

Discussion

Major efforts to identify new drugs to treat the adult stage (macrofilariae) of Onchocerca volvulus have led to the discovery of novel anti-Wolbachia compounds. These studies demonstrated that their respective anti-Wolbachia compounds significantly reduced Wolbachia titers in worms from animals treated with short courses of quinazolines and Tylosin analogs after 16–18 weeks post-first dose [26,27,29,40,50]. In addition, a one-week combination treatment of rifampicin and albendazole resulted in a greater than 99% reduction in Wolbachia levels in B. malayi infected SCID mice [28]. Clinical trials further revealed that albendazole either alone or in combination with antibiotics dramatically reduced Wolbachia levels [51,52]. With the recent discoveries of new anti-wolbachial drugs, we explored the Wolbachia/worm relationship in vivo to better understand the outcomes of antibiotic exposure. The aim of this study was to determine the longer-term effects of rifampicin on female worm survival and fecundity and Wolbachia titers using the Brugia pahangi infected jird model.

Although macrofilaricidal effects were not observed following treatment with rifampicin, Wolbachia titers and female fecundity were significantly affected 1 week post-first dose. Wolbachia titers were reduced by >95% in female worms from rifampicin treated animals at the 1-week timepoint compared to those of worms from vehicle animals. The extensive reduction of Wolbachia titers in adult worms has been previously described in other rodent studies with filarial worms, but the studies were terminated 16–18 weeks post-treatment [26,27,29,40,50], and the long-term effects of antibiotic treatment on filarial worms and their Wolbachia were not evaluated in these models.

Our present long-term study showed that even though Wolbachia were almost completely eliminated at early timepoints, Wolbachia rebounded to levels similar to those of control worms 8 months after rifampicin dosing. Female worms recovered from this timepoint were also fully reproductive, similar to female worms from the control group. Thus, despite qPCR data that showed a >95% reduction in Wolbachia titers, Wolbachia rebounded and returned to levels comparable to those of control worms, suggesting that this level of elimination is not sufficient to ensure permanent elimination and permanent female sterility. This finding parallels the rebound of Wolbachia titers in O. ochengi from cattle treated with a short, intensive regimen of oxytetracycline (10 mg/kg QD for 14 days) reported by Gilbert et al. [32], and suggests that this jird model is a suitable alternative for long-term studies (up to 8 months) in situations where cattle models cannot be conducted.

The dosage of rifampicin used in the present study was not likely suboptimal since previous studies with B. malayi infected SCID mice showed that 25 mg/kg rifampicin twice a day for 7 days resulted in reductions in Wolbachia comparable to the clinical dosage of doxycycline that caused >90% Wolbachia depletion [25]. While we saw the expected reduction in Wolbachia at early timepoints, the reduction was not sustained, and Wolbachia rebounded. Comparison of the genetic profiles of Wolbachia genomes from the early and late timepoints revealed that despite the population bottleneck and recovery, no genetic changes occurred in Wolbachia that could account for the rebound.

The significance of the rebound in this bacteria/worm symbiosis was evident when low titers of Wolbachia, which initially led to the reduction of late-stage embryos and microfilariae at 1- and 6- weeks, was later followed by the return of developing embryos and microfilariae released by female worms. This recovery back to control levels was commensurate with Wolbachia rebound, in line with evidence showing the dependence of female worms on Wolbachia to maintain their reproductive output [21,32,49,5355].

Direct visualization of worm ovaries using fluorescence confocal microscopy revealed two distinct populations of Wolbachia: “clusters” of Wolbachia that were found within the female ovaries and “peripheral” Wolbachia that were found surrounding the clusters. For the Wolbachia within clusters, we found no significant differences in bacterial cluster amount, size, or density between rifampicin and control treatments, suggesting that these clusters do not respond to antibiotics, at least at the dosages used in this study. In contrast, we found that rifampicin significantly reduced the peripheral Wolbachia surrounding the clusters at 6 weeks. To our knowledge, these clusters have never before been identified in any Wolbachia-nematode system, and we believe that the clusters could serve as a reservoir of bacteria that can repopulate the germline tissue after antibiotic treatment.

Since genome sequencing of the rebounded Wolbachia showed there were no gene changes that could account for the persistence, we postulate that the clusters are privileged sites in which Wolbachia persist in a low or inactive metabolic state, similar to what occurs with intracellular Toxoplasma gondii bradyzoites [56,57] and pathogenic intracellular bacteria including Mycobacterium tuberculosis, Treponema pallidum, Chlamydia spp. and Salmonella enterica which can cause persistent and latent infections [5866]. Interestingly, bacterial toxin-antitoxin (TA) genes of the RelEB family thought to cause persister cell formation in insect-associated Wolbachia, were absent in Wolbachia from filarial nematodes, suggesting that TAs may not be involved in persister formation of Wolbachia within the clusters and that other mechanisms are likely at play [6770].

Although the molecular mechanisms of persister formation is not known in the Wolbachia-worm relationship, Wolbachia may respond in some manner to their low numbers and repopulate the ovarian tissues by moving from the clusters to the peripheral areas within the ovaries or by migrating into the worm’s pseudocoelom [46,71] and back into the ovaries, thereby ensuring their vertical transmission for future generations of microfilariae.

Further studies to define the nature of these clusters and wolbachial persistence will yield new information on the cell biology of this bacteria-worm symbiosis and may reveal similar strategies used by various pathogens that allow them to persist and remain latent within their hosts.

Conclusion

We describe the effects of rifampicin treatment on the Wolbachia-Brugia pahangi relationship over an 8-month period in a rodent model. Wolbachia numbers were significantly reduced after initial treatment but subsequently rebounded along with a corresponding return of embryogenesis and fecundity in female worms. This in vivo B. pahangi/jird model serves as a useful tool to evaluate the long-term effects of antibiotics on Wolbachia depletion and female worm fecundity and provides information that may impact the clinical use of antibiotics to treat filarial diseases.

This study also provides insight into the Wolbachia-worm relationship with the discovery of two different populations of Wolbachia within the ovaries of these filarial worms: clusters of Wolbachia and peripheral Wolbachia, the former of which may account for the rebound of Wolbachia following antibiotic treatment. To our knowledge, clusters of Wolbachia have never before been identified in any Wolbachia-nematode system and may represent sequestered populations of this endosymbiont within Brugia pahangi ovaries.

Methods

Ethics statement

All animal studies were performed under the University of California, San Francisco Institutional Animal Care and Use Committee (IACUC) approvals AN109629-03 and AN173847-02 and adhered to the guidelines set forth in the NIH Guide for the Care and Use of Laboratory Animals and the USDA Animal Care Policies.

Animal infections

For dosing studies on adult worms, male Mongolian jirds 50–60 grams, 5–7 weeks in age (Meriones unguiculatus, Charles River Laboratories International, Inc., Wilmington, MA) were injected intraperitoneally (IP) with third-larval stage Brugia pahangi (University of Missouri-Columbia) and treated 3 months later when larvae developed into adult worms.

Drug dosages

Rifampicin (Research Products International Corp., Prospect, IL) was dissolved in 55% polyethylene glycol 400 (Sigma), 25% propylene glycol (Sigma), 20% water at a concentration of 5 mg/mL, and animals were given oral doses of 25 mg/kg twice a day for 7 days. We selected the dosage 25 mg/kg BID for 7 days based on findings from Aljayyoussi et al, 2017 which reported rifampicin dosages of 15 mg/kg QD for 7 days, 35 mg/kg QD for 7 days, or 25 mg/kg BID for 7 days resulted in reductions in Wolbachia of 97.7%, 98.2% and 99.5%, respectively [25]. They found 25 mg/kg BID x 7 days to be superior to doxycycline 25 mg/kg BID treatment for four weeks (P>0.0001) and not significantly inferior from doxycycline 25 mg/kg BID treatment for six weeks [25].

PK analyses

To determine the level of exposure of rifampicin in treated animals, blood was collected from the saphenous vein in Am-heparinized tubes and centrifuged at 2,000 x g for 15 min at 4°C. Blood was collected 0.5 hour, 1 hour, 3 hours and 6 hours post-first dose. The second dose was given 8 hours post-first dose and blood was collected 24 hours post-first dose. Plasma was collected and stored at -80°C prior to shipment to Integrated Analytical Solutions (Berkeley, CA) for plasma analysis (S2 Fig). Calibration standards, QC samples and study samples were processed for LC/MS/MS analysis by precipitating 10 μL of each sample with 3 volumes of ice cold Internal Standard Solution (acetonitrile containing 50 ng/mL dextromethorphan). The precipitated samples were centrifuged at 6100g for 30 min and an aliquot of each supernatant was transferred to an auto sampler plate and diluted in water with 2 volumes of 0.2% formic acid.

Necropsies

Vehicle and rifampicin treated jirds were necropsied 1 week, 6 weeks, 17 weeks and 8 months post-first dose. Animals were dissected and peritoneal cavities were washed with 100 mL of PBS to collect adult worms and mf that had been released from female worms. Adult worms were separated by sex, counted and processed for subsequent analyses. Mf from the peritoneal cavity were quantified by mixing peritoneal wash 9:1 (v/v) with 0.04% methylene blue:water and counted using an inverted microscope. Data from each timepoint are from three replicate studies, except for the 17-week timepoint, in which the data are from one set. All animals were euthanized at their intended timepoints and no adverse events were observed. However, one animal had ascites, but was otherwise healthy at the 8-month timepoint.

qPCR analysis of Wolbachia in adult Brugia pahangi

Adult worms collected during necropsies were snap-frozen in a dry-ice and ethanol bath prior to storage at -80° C. gDNA was extracted from individual female worms or from 4–25 male worms using a DNEasy Blood & Tissue Kit (QIAGEN) according to the manufacturer’s instructions. Genomic DNA was quantified using a NanoDrop Onec (Thermo Fisher Scientific) and qPCR was performed using a Geneoecopia 2x All-in-One Master Mix (Cat #QP001-01) in a Bio-Rad CFX Connect RT-PCR thermocycler. The single copy gene, Wolbachia surface protein (wsp), was used to quantify Wolbachia titers and the single copy gene, glutathione-S-transferase (gst), was used to quantify Brugia titers following the protocol of McGarry et al. [37]. Primers used for qPCR were based on wsp forward: 5’-CCCTGCAAAGGCACAAGTTATTG-3’; wsp reverse: 5’-CGAGCTCCAGCAAAGAGTTTAATTT-3’; gst foward: 5’-GAGACACCTTGCTCGCAAAC-3’; gst reverse: 5’-ATCACGGACGCCTTCACAG-3’. For gst amplification the following cycles were used: 95°C for 15 minutes, followed by 36 cycles of denaturation at 94°C for 15 seconds, annealing at 55° C for 30 seconds and elongation at 72° C for 30 seconds. Melting curve analysis was conducted by heating to 95°C for 1 minute, annealing at 55°C for 30 seconds and heating to 97°C. For wsp amplification: samples were heated at 95° C for 15 minutes, followed by 40 cycles of denaturation at 94°C for 10 seconds, annealing at 55°C for 20 seconds and elongation at 72°C for 15 seconds. Melting curve analysis was conducted by heating to 95°C for 1 minute, annealing at 55°C for 30 seconds and heating to 97°C.

Wolbachia genome sequencing

Total genomic DNA was extracted from Brugia pahangi worms recovered from 1-week and 8-month timepoints using DNeasy Blood & Tissue Kit (QIAGEN) following the manufacturer’s protocol and quantified using Qubit (Invitrogen). Genomic DNA was subjected to hybridization probe-capture (adaptation of the protocol of Geniez et al. and protocol of Lefoulon et al.) to enrich for Wolbachia DNA using biotinylated probe-baits and magnetic streptavidin beads [41,72]. Prior to capture, genomic DNA was sheared using NEBNext FSII for 30 min at 37° and ligated to NimbleGen SeqCap adapters (NEBNext Ultra II kit), followed by AMPure bead purification (0.9X), PCR amplification and purification through AMPure beads (0.9X). The barcoded samples were then pooled (~400 ng per sample) and hybridization of DNA with Wolbachia specific EZ library probes was performed according to SeqCap EZ HyperCap protocol v1.0 (NimbleGen). The captured DNA library was amplified by PCR, purified using AMPure bead (0.9X), and subjected to PacBio circular consensus sequencing (CCS). A reference wBp genome was assembled with Canu v1.9 [73] from the control sample (1-week vehicle group) after the CCS reads were trimmed to remove residual adapter sequences using seqtk. To minimize assembly errors due to the presence of B. pahangi sequences derived from nuclear Wolbachia transfers (nuwts), a draft assembly was first generated using all available reads, and then reads (longer than 3 kb) that mapped to the assembly without clipping were collected using minimap v2.17 [74] and assembled to produce a second draft assembly, which was then circularized using Circlator [75]. Assembly errors were further corrected through manual curation and Pilon v1.23 [76]. Genome annotation was performed using PGAP [77] and DFAST v1.2.4 [78]. Genetic variants (SNPs and indels) were called across samples by DeepVariant [79] after non-clipped CCS reads were aligned to the wBp reference genome using minimap2. The depth-of-coverage patterns of clipped reads (that span the junctions between nuwts and B. pahangi DNA) along the Wolbachia genome were exploited to infer the location of regions that show sequence similarity to nuwts using samtools [80] and sequana_coverage v0.7.1 [81]. Using VCFtools v0.1.17 [82], false-positive variants occurring within these putative nuwts regions were filtered and excluded from further analyses. Finally, SnpEff v4.3 [83] was used to annotate and predict the effects of the remaining variants. The statistical significance of allele frequency differences was determined with Fisher's exact test.

Embryograms and microfilariae overnight shed

Individual adult female worms (n = 12–24 females per treatment group) from vehicle and rifampicin treated jirds were maintained overnight in 24-well plates with 500 μL of RPMI-1640, 25mM HEPES, 5% heat inactivated-FBS, and 1x Antibiotic/Antimycotic. Mf that were released from individual females after 18 hours were removed from the wells and counted. For the embryogram analyses, individual adult female worms previously frozen in 0.5mL of 0.1% PBS-Triton X-100 (Sigma) were homogenized using a glass pestle to disrupt the cuticle and expose the reproductive structures. Developing stages from the ovaries and uteri (oocyte, early morula, late morula, pre-mf, pretzel mf and stretched mf, degenerated embryos) were assessed in a blinded fashion for their developmental stages using an inverted microscope and hemocytometer. A minimum of 100 developmental stages were counted from each female and relative proportions were used to determine the means and standard deviations. n = 6–9 females per treatment group per timepoint [45].

Fluorescence imaging of female worm ovaries

Female B. pahangi worms removed from jirds from each group were frozen and shipped to UC Santa Cruz for fluorescence analysis. Frozen worms were thawed at room temperature, immediately fixed in 3.2% paraformaldehyde for 25 minutes and rinsed twice in PBST (PBS plus 0.1% Triton-X100). Individual female uteri/ovaries were dissected from fixed tissue and incubated overnight in RNAse A (10mg/mL) in PBST following similar protocols described by Landmann et al., Serbus et al. and Foray et al. [4549]. Tissues were then stained with propidium iodide (PI) (1mg/mL diluted 100X in PBST) for 30 seconds, rinsed twice in PBST and then mounted in DAPI Vectashield mounting medium (Vector Labs). The distal tips of the ovarian tissue were imaged on a Leica SP5 confocal microscope and single images were taken at the mid-plane of the ovarian tissue for each distal tip. To measure the average Wolbachia titer of each distal tip, Wolbachia puncta were counted by hand using the Cell Counter tool in FIJI; the number of puncta was divided by the area of the tissue in each image (puncta/μm2).

Statistical analyses

Data were first tested for normality using the Shapiro-Wilk test of normality. When data did not pass the normality test, a Mann-Whitney U test was conducted and when data did pass the normality test, a Student’s t-test was used. All significance levels were determined as compared to the vehicle worms at the same timepoint. Individual percent reductions of Wolbachia were calculated for each worm using the wsp copy number normalized to the Brugia gst copy number [28,29,3840]. Wsp/gst ratios were calculated for each timepoint by subtracting the wsp/gst ratios of treated worms from the mean wsp/gst ratio of vehicle worms and then dividing by the mean wsp/gst ratio of vehicle worms for each respective timepoint. The means were then calculated to determine the average percent reduction. All statistical analyses were determined using Prism 8 version 8.2.0 (272).

Supporting information

S1 Fig. Rifampicin treatment of Brugia pahangi-infected jirds does not reduce worm burden significantly at any timepoint.

(A) Number of total worms recovered from the peritoneal cavity of B. pahangi-infected jirds 1-week, 6-weeks, 17-weeks and 8-months post-first dose. (B) Number of female worms recovered from the peritoneal cavity of B. pahangi-infected jirds 1-week, 6-weeks, 17-weeks and 8-months post-first dose. (C) Number of male worms recovered from the peritoneal cavity of B. pahangi-infected jirds 1-week, 6-weeks, 17-weeks and 8-months post-first dose. Data is presented as median ± 95% CI. n = 2–9 jirds per treatment group per timepoint.

https://doi.org/10.1371/journal.ppat.1008623.s001

(PDF)

S2 Fig. Plasma concentrations peak 3 hours post-rifampicin treatment of Brugia pahangi-infected jirds.

Plasma samples were collected at 0.5, 1, 3, 6 and 24 hours post-first dose from B. pahangi infected jirds treated with oral doses of rifampicin 25 mg/kg twice a day for 7 days. Analyses showed that plasma concentrations peaked at 3 hours post-first dose with a Cmax of 5.17x103 ng/mL. n = 3 jirds for all timepoints except n = 2 at 6 hours and n = 4 at 24 hours.

https://doi.org/10.1371/journal.ppat.1008623.s002

(PDF)

S1 Table. Results of Wolbachia titers from 3 experimental replicates after rifampicin treatment.

qPCR analyses used worms from 3 separate cohorts of animals with data from two replicate experiments for each timepoint, except for the 17-week timepoint. Data are presented as mean percent reductions based on control worms.

https://doi.org/10.1371/journal.ppat.1008623.s003

(PDF)

S2 Table. PacBio sequencing of Wolbachia using hybridization probe-capture.

https://doi.org/10.1371/journal.ppat.1008623.s004

(PDF)

S3 Table. Genetic variants identified in Wolbachia after rifampicin treatment.

https://doi.org/10.1371/journal.ppat.1008623.s005

(PDF)

S4 Table. Female worms recovered from jirds treated 6 weeks post-first dose had significantly higher numbers of degenerated embryos compared to those from the vehicle group.

The number in each cell is the mean of each embryonic stage ± the standard deviation.

https://doi.org/10.1371/journal.ppat.1008623.s006

(PDF)

Acknowledgments

We thank Richard Spear for his generous support of the Sullivan lab.

References

  1. 1. James SL, Abate D, Abate KH, Abay SM, Abbafati C, Abbasi N, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 354 Diseases and Injuries for 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018; 1789–1858. pmid:30496104
  2. 2. Taylor MJ, Hoerauf A, Bockarie M. Lymphatic filariasis and onchocerciasis. Lancet. 2010;376: 1175–1185. pmid:20739055
  3. 3. Abdulmalik J, Nwefoh E, Obindo J, Dakwak S, Ayobola M, Umaru J, et al. Emotional difficulties and experiences of stigma among persons with lymphatic filariasis in Plateau state, Nigeria. Health Hum Rights. 2018;20: 27–40. pmid:30008550
  4. 4. Molyneux DH, Bradley M, Hoerauf A, Kyelem D, Taylor MJ. Mass drug treatment for lymphatic filariasis and onchocerciasis. Trends Parasitol. 2003;19: 516–522. pmid:14580963
  5. 5. Ottesen EA, Hooper PJ, Bradley M, Biswas G. The global programme to eliminate lymphatic filariasis: Health impact after 8 years. PLoS Negl Trop Dis. 2008;2. pmid:18841205
  6. 6. World Health Organization. Global Programme to Eliminate Lymphatic Filariasis: Progress Report 2000–2009 and Strategic Plan 2010–2020. 2010.
  7. 7. Chu BK, Hooper PJ, Bradley MH, McFarland DA, Ottesen EA. The economic benefits resulting from the first 8 years of the Global Programme to eliminate Lymphatic Filariasis (2000–2007). PLoS Negl Trop Dis. 2010;4. pmid:20532228
  8. 8. Hoerauf A, Pfarr K, Mand S, Debrah AY, Specht S. Filariasis in Africa-treatment challenges and prospects. Clin Microbiol Infect. 2011;17: 977–985. pmid:21722251
  9. 9. Molyneux DH, Taylor MJ. Current status and future prospects of the global lymphatic filariasis programme. Curr Opin Infect Dis. 2001;14: 155–159. pmid:11979126
  10. 10. Molyneux DH, Hopkins A, Bradley MH, Kelly-Hope LA. Multidimensional complexities of filariasis control in an era of large-scale mass drug administration programmes: a can of worms. Parasit Vectors. 2014;7: 363. pmid:25128408
  11. 11. Turner HC, Walker M, Churcher TS, Osei-Atweneboana MY, Biritwum NK, Hopkins A, et al. Reaching the London declaration on neglected tropical diseases goals for onchocerciasis: An economic evaluation of increasing the frequency of ivermectin treatment in Africa. Clin Infect Dis. 2014;59: 923–932. pmid:24944228
  12. 12. Kim YE, Remme JHF, Steinmann P, Stolk WA, Roungou JB, Tediosi F. Control, Elimination, and Eradication of River Blindness: Scenarios, Timelines, and Ivermectin Treatment Needs in Africa. PLoS Negl Trop Dis. 2015;9: 1–19. pmid:25860569
  13. 13. Basáñez MG, Walker M, Hamley JI, Milton P, Fronterre C, de Vlas SJ, et al. The World Health Organization 2030 goals for onchocerciasis: Insights and perspectives from mathematical modelling. Gates Open Res. 2019;3. pmid:31723729
  14. 14. Davis EL, Vlas SJ de, Fronterre C, Hollingsworth TD, Kontoroupis P, Michael E, et al. The roadmap towards elimination of lymphatic filariasis by 2030: insights from quantitative and mathematical modelling. Gates Open Res. 2019;3. pmid:31728440
  15. 15. World Health Organization. WHO launches global consultations for a new roadmap on neglected tropical diseases. 2019.
  16. 16. Gardon J, Gardon-Wendel N, Demanga-Ngangue , Kamgno J, Chippaux JP, Boussinesq M. Serious reactions after mass treatment of onchocerciasis with ivermectin in an area endemic for Loa loa infection. Lancet. 1997;350: 18–22. pmid:9217715
  17. 17. Boussinesq M, Gardon J, Gardon-Wendel N, Chippaux J-P. Clinical picture, epidemiology and outcome of Loa-associated serious adverse events related to mass ivermectin treatment of onchocerciasis in Cameroon. Filaria J. 2003;2 Suppl 1: S4. pmid:14975061
  18. 18. Bandi C, Anderson TJ, Genchi C, Blaxter ML. Phylogeny of Wolbachia in filarial nematodes. Proc R Soc London Biol Sci. 1998;265: 2407–2413. pmid:9921679
  19. 19. Slatko BE, Taylor MJ, Foster JM. The Wolbachia endosymbiont as an anti-filarial nematode target. Symbiosis. 2010;51: 55–65. pmid:20730111
  20. 20. Slatko BE, Luck AN, Dobson SL, Foster JM. Wolbachia endosymbionts and human disease control. Mol Biochem Parasitol. 2014;195: 88–95. pmid:25046729
  21. 21. Hoerauf A, Volkmann L, Hamelmann C, Adjei O, Autenrieth IB, Fleischer B, et al. Endosymbiotic bacteria in worms as targets for a novel chemotherapy in filariasis. Lancet. 2000;355: 1242–1243. pmid:10770311
  22. 22. Taylor MJ, Hoerauf A, Townson S, Slatko BE, Ward SA. Anti-Wolbachia drug discovery and development: safe macrofilaricides for onchocerciasis and lymphatic filariasis. Parasitology. 2014;141: 119–127. pmid:23866958
  23. 23. Taylor MJ, Makunde WH, McGarry HF, Turner JD, Mand S, Hoerauf A. Macrofilaricidal activity after doxycycline treatment of Wuchereria bancrofti: a double-blind, randomised placebo-controlled trial. Lancet. 2005;365: 2116–2121. pmid:15964448
  24. 24. Debrah AY, Specht S, Klarmann-schulz U, Batsa L, Mand S, Marfo-Debrekyei Y, et al. Doxycycline Leads to Sterility and Enhanced Killing of Female Onchocerca volvulus Worms in an Area With Persistent Microfilaridermia After Repeated Ivermectin Treatment: A Randomized, Placebo-Controlled, Double-Blind Trial. Clin Infect Dis. 2015;61: 517–526. pmid:25948064
  25. 25. Aljayyoussi G, Tyrer HE, Ford L, Sjoberg H, Pionnier N, Waterhouse D, et al. Short-course, high-dose rifampicin achieves Wolbachia depletion predictive of curative outcomes in preclinical models of lymphatic filariasis and onchocerciasis. Sci Rep. 2017;7: 1–12.
  26. 26. Bakowski MA, Shiroodi RK, Liu R, Olejniczak J, Yang B, Gagaring K, et al. Discovery of short-course antiwolbachial quinazolines for elimination of filarial worm infections. Sci Transl Med. 2019;11. pmid:31068442
  27. 27. Hübner MP, Koschel M, Struever D, Nikolov V, Frohberger SJ, Ehrens A, et al. In vivo kinetics of Wolbachia depletion by ABBV-4083 in L. sigmodontis adult worms and microfilariae. PLoS Negl Trop Dis. 2019;13: 1–19. pmid:31381563
  28. 28. Turner JD, Sharma R, Al Jayoussi G, Tyrer HE, Gamble J, Hayward L, et al. Albendazole and antibiotics synergize to deliver short-course anti-Wolbachia curative treatments in preclinical models of filariasis. Proc Natl Acad Sci. 2017;114: E9712–E9721. pmid:29078351
  29. 29. Hübner MP, Gunderson E, Vogel I, Bulman CA, Lim KC, Koschel M, et al. Short-course quinazoline drug treatments are effective in the Litomosoides sigmodontis and Brugia pahangi jird models. Int J Parasitol Drugs Drug Resist. 2020;12: 18–27. pmid:31869759
  30. 30. Langworthy NG, Renz A, Mackenstedt U, Henkle-Dührsen K, De C Bronsvoort MB, Tanya VN, et al. Macrofilaricidal activity of tetracycline against the filarial nematode Onchocerca ochengi: Elimination of Wolbachia precedes worm death and suggests a dependent relationship. Proc R Soc B Biol Sci. 2000;267: 1063–1069. pmid:10885510
  31. 31. Trees AJ, Graham SP, Renz A, Bianco AE, Tanya V. Onchocerca ochengi infections in cattle as a model for human onchocerciasis: Recent developments. Parasitology. 2000;120: 133–142. pmid:10874716
  32. 32. Gilbert J, Nfon CK, Makepeace BL, Njongmeta LM, Hastings IM, Pfarr KM, et al. Antibiotic Chemotherapy of Onchocerciasis: In a Bovine Model, Killing of Adult Parasites Requires a Sustained Depletion of Endosymbiotic Bacteria (Wolbachia Species). J Infect Dis. 2005;192: 1483–1493. pmid:16170768
  33. 33. Nfon CK, Makepeace BL, Njongmeta LM, Tanya VN, Bain O, Trees AJ. Eosinophils contribute to killing of adult Onchocerca ochengi within onchocercomata following elimination of Wolbachia. Microbes Infect. 2006;8: 2698–2705. pmid:16962357
  34. 34. Nfon CK, Makepeace BL, Njongmeta LM, Tanya VN, Trees AJ. Lack of resistance after re-exposure of cattle cured of Onchocerca ochengi infection with oxytetracycline. Am J Trop Med Hyg. 2007;76: 67–72. pmid:17255232
  35. 35. Bah GS, Ward EL, Srivastava A, Trees AJ, Tanya VN, Makepeace BL. Efficacy of three-week oxytetracycline or rifampin monotherapy compared with a combination regimen against the filarial nematode Onchocerca ochengi. Antimicrob Agents Chemother. 2014;58: 801–810. pmid:24247133
  36. 36. Bah GS, Tanya VN, Makepeace BL. Immunotherapy with mutated onchocystatin fails to enhance the efficacy of a sub-lethal oxytetracycline regimen against Onchocerca ochengi. Vet Parasitol. 2015;212: 25–34. pmid:26100152
  37. 37. McGarry HF, Egerton GL, Taylor MJ. Population dynamics of Wolbachia bacterial endosymbionts in Brugia malayi. Mol Biochem Parasitol. 2004;135: 57–67. pmid:15287587
  38. 38. Halliday A, Guimaraes AF, Tyrer HE, Metuge HM, Patrick CNW, Arnaud K-OJ, et al. A murine macrofilaricide pre-clinical screening model for onchocerciasis and lymphatic filariasis. Parasit Vectors. 2014;7: 472. pmid:25338621
  39. 39. Johnston KL, Ford L, Umareddy I, Townson S, Specht S, Pfarr K, et al. Repurposing of approved drugs from the human pharmacopoeia to target Wolbachia endosymbionts of onchocerciasis and lymphatic filariasis. Int J Parasitol Drugs Drug Resist. 2014;4: 278–286. pmid:25516838
  40. 40. Taylor MJ, Von Geldern TW, Ford L, Hubner MP, Marsh K, Johnston KL, et al. Preclinical development of an oral anti-Wolbachia macrolide drug for the treatment of lymphatic filariasis and onchocerciasis. Sci Transl Med. 2019;11: 1–11. pmid:30867321
  41. 41. Lefoulon E, Vaisman N, Frydman HM, Sun L, Foster JM, Slatko BE. Large Enriched Fragment Targeted Sequencing (LEFT-SEQ) Applied to Capture of Wolbachia Genomes. Sci Rep. 2019; 1–10.
  42. 42. Hotopp JCD, Slatko BE, Foster JM. Targeted Enrichment and Sequencing of Recent Endosymbiont-Host Lateral Gene Transfers. Sci Rep. 2017;7: 1–10.
  43. 43. Wernegreen JJ, Kauppinen SN, Degnan PH. Slip into something more functional: Selection maintains ancient frameshifts in homopolymeric sequences. Mol Biol Evol. 2010;27: 833–839. pmid:19955479
  44. 44. Fang H, Wu Y, Narzisi G, O’Rawe JA, Barrón LTJ, Rosenbaum J, et al. Reducing INDEL calling errors in whole genome and exome sequencing data. Genome Med. 2014;6: 1–17.
  45. 45. Landmann F, Foster JM, Slatko B, Sullivan W. Asymmetric wolbachia segregation during Early Brugia malayi embryogenesis determines its distribution in adult host tissues. PLoS Negl Trop Dis. 2010;4. pmid:20689574
  46. 46. Landmann F, Bain O, Martin C, Uni S, Taylor MJ, Sullivan W. Both asymmetric mitotic segregation and cell-to-cell invasion are required for stable germline transmission of Wolbachia in filarial nematodes. Biol Open. 2012;1: 536–547. pmid:23213446
  47. 47. Landmann F, Foster JM, Slatko BE, Sullivan W. Efficient in vitro RNA interference and immunofluorescence-based phenotype analysis in a human parasitic nematode, Brugia malayi. Parasites and Vectors. 2012;5: 1–8.
  48. 48. Serbus LR, Landmann F, Bray WM, White PM, Ruybal J, Lokey RS, et al. A Cell-Based Screen Reveals that the Albendazole Metabolite, Albendazole Sulfone, Targets Wolbachia. PLoS Pathog. 2012;8. pmid:23028321
  49. 49. Foray V, Pérez-Jiménez MM, Fattouh N, Landmann F. Wolbachia Control Stem Cell Behavior and Stimulate Germline Proliferation in Filarial Nematodes. Dev Cell. 2018;45: 198–211.e3. pmid:29689195
  50. 50. Hong WD, Benayoud F, Nixon GL, Ford L, Johnston KL, Clare RH, et al. AWZ1066S, a highly specific anti-Wolbachia drug candidate for a short-course treatment of filariasis. Proc Natl Acad Sci. 2019;116: 1414 LP– 1419. pmid:30617067
  51. 51. Klarmann-Schulz U, Specht S, Debrah AY, Batsa L, Ayisi-Boateng NK, Osei-Mensah J, et al. Comparison of Doxycycline, Minocycline, Doxycycline plus Albendazole and Albendazole Alone in Their Efficacy against Onchocerciasis in a Randomized, Open-Label, Pilot Trial. PLoS Negl Trop Dis. 2014;11: e0005156. pmid:28056021
  52. 52. Gayen P, Nayak A, Saini P, Mukherjee N, Maitra S, Sarkar P, et al. A double-blind controlled field trial of doxycycline and albendazole in combination for the treatment of bancroftian filariasis in India. Acta Trop. 2013;125: 150–156. pmid:23123345
  53. 53. Volkmann L, Fischer K, Taylor M, Hoerauf A. Antibiotic therapy in murine filariasis (Litomosoides sigmodontis): Comparative effects of doxycycline and rifampicin on Wolbachia and filarial viability. Trop Med Int Heal. 2003;8: 392–401. pmid:12753632
  54. 54. Hoerauf A, Nissen-Pähle K, Schmetz C, Henkle-Dührsen K, Blaxter ML, Büttner DW, et al. Tetracycline therapy targets intracellular bacteria in the filarial nematode Litomosoides sigmodontis and results in filarial infertility. J Clin Invest. 1999;103: 11–18. pmid:9884329
  55. 55. Townson S, Tagboto S, McGarry HF, Egerton GL, Taylor MJ. Onchocerca parasites and Wolbachia endosymbionts: Evaluation of a spectrum of antibiotic types for activity against Onchocerca gutturosa in vitro. Filaria J. 2006;5: 1–9.
  56. 56. Weiss LM, Kim K. The development and biology of bradyzoites of Toxoplasma gondii. Front Biosci. 2000;5. pmid:10762601
  57. 57. Sullivan WJ, Jeffers V. Mechanisms of Toxoplasma gondii persistence and latency. FEMS Microbiol Rev. 2012;36: 717–733. pmid:22091606
  58. 58. Torrey HL, Keren I, Via LE, Lee JS, Lewis K. High persister mutants in mycobacterium tuberculosis. PLoS One. 2016;11: 1–28. pmid:27176494
  59. 59. Cameron DR, Shan Y, Zalis EA, Isabella V, Lewis K. A genetic determinant of persister cell formation in bacterial pathogens. J Bacteriol. 2018;200: 1–11. pmid:29941425
  60. 60. Prasetyoputri A, Jarrad AM, Cooper MA, Blaskovich MAT. The Eagle Effect and Antibiotic-Induced Persistence: Two Sides of the Same Coin? Trends Microbiol. 2019;27: 339–354. pmid:30448198
  61. 61. Rittershaus E, Baek S, Sassetti C. The normalcy of dormancy. Cell Host Microbe. 2013;13: 643–651. pmid:23768489
  62. 62. Ehrt S, Schnappinger D, Rhee KY. Metabolic principles of persistence and pathogenicity in Mycobacterium tuberculosis. Nat Rev Microbiol. 2018;16: 496–507. pmid:29691481
  63. 63. Singh AE, Romanowski B. Syphilis: Review with emphasis on clinical, epidemiologic, and some biologic features. Clin Microbiol Rev. 1999;12: 187–209. pmid:10194456
  64. 64. Panzetta ME, Valdivia RH, Saka HA. Chlamydia persistence: A survival strategy to evade antimicrobial effects in-vitro and in-vivo. Front Microbiol. 2018;9: 1–11.
  65. 65. Monack DM, Mueller A, Falkow S. Persistent bacterial infections: The interface of the pathogen and the host immune system. Nat Rev Microbiol. 2004;2: 747–765. pmid:15372085
  66. 66. Fisher RA, Gollan B, Helaine S. Persistent bacterial infections and persister cells. Nat Rev Microbiol. 2017;15: 453–464. pmid:28529326
  67. 67. Singhal K, Mohanty S. Comparative genomics reveals the presence of putative toxin–antitoxin system in Wolbachia genomes. Mol Genet Genomics. 2018;293: 525–540. pmid:29214346
  68. 68. Helaine S, Cheverton AM, Watson KG, Faure LM, Matthews SA, Holden DW. Internalization of Salmonella by Macrophages Induces Formation of Nonreplicating Persisters. Science. 2014;343: 204–208. pmid:24408438
  69. 69. Harms A, Brodersen DE, Mitarai N, Gerdes K. Toxins, Targets, and Triggers: An Overview of Toxin-Antitoxin Biology. Mol Cell. 2018;70: 768–784. pmid:29398446
  70. 70. Fallon AM. Computational evidence for antitoxins associated with RelE/ParE, RatA, Fic, and AbiEii-family toxins in Wolbachia genomes. Mol Genet Genomics. 2020. pmid:32189066
  71. 71. Fischer K, Beatty WL, Weil GJ, Fischer PU. High pressure freezing/freeze substitution fixation improves the ultrastructural assessment of Wolbachia endosymbiont—Filarial nematode host interaction. PLoS One. 2014;9: 16–19. pmid:24466066
  72. 72. Geniez S, Foster JM, Kumar S, Moumen B, Leproust E, Hardy O, et al. Targeted genome enrichment for efficient purification of endosymbiont DNA from host DNA. Symbiosis. 2012;58: 201–207. pmid:23482460
  73. 73. Koren S, Walenz BP, Konstantin B, Miller JR, Bergman NH, Phillippy AM. Canu: scalable and accurate long-read assembly via adaptive k-mer weighting and repeat separation. Genome Res. 2017;27: 722–736. pmid:28298431
  74. 74. Li H. Minimap2: Pairwise alignment for nucleotide sequences. Bioinformatics. 2018;34: 3094–3100. pmid:29750242
  75. 75. Hunt M, Silva N De, Otto TD, Parkhill J, Keane JA, Harris SR. Circlator: Automated circularization of genome assemblies using long sequencing reads. Genome Biol. 2015;16: 1–10.
  76. 76. Walker BJ, Abeel T, Shea T, Priest M, Abouelliel A, Sakthikumar S, et al. Pilon: An integrated tool for comprehensive microbial variant detection and genome assembly improvement. PLoS One. 2014;9. pmid:25409509
  77. 77. Tatusova T, Dicuccio M, Badretdin A, Chetvernin V, Nawrocki EP, Zaslavsky L, et al. NCBI prokaryotic genome annotation pipeline. Nucleic Acids Res. 2016;44: 6614–6624. pmid:27342282
  78. 78. Tanizawa Y, Fujisawa T, Nakamura Y. DFAST: A flexible prokaryotic genome annotation pipeline for faster genome publication. Bioinformatics. 2018;34: 1037–1039. pmid:29106469
  79. 79. Poplin R, Chang PC, Alexander D, Schwartz S, Colthurst T, Ku A, et al. A universal snp and small-indel variant caller using deep neural networks. Nat Biotechnol. 2018;36: 983. pmid:30247488
  80. 80. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25: 2078–2079. pmid:19505943
  81. 81. Desvillechabrol D, Bouchier C, Kennedy S, Cokelaer T. Sequana coverage: Detection and characterization of genomic variations using running median and mixture models. Gigascience. 2018;7: 1–13. pmid:30192951
  82. 82. Danecek P, Auton A, Abecasis G, Albers CA, Banks E, DePristo MA, et al. The variant call format and VCFtools. Bioinformatics. 2011;27: 2156–2158. pmid:21653522
  83. 83. Cingolani P, Platts A, Wang LLL, Coon M, Nguyen T, Wang LLL, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6: 80–92. pmid:22728672