Factors Regulating Murine Tuft cell Hyperplasia in the Small Intestine during Infection with Hymenolepis Diminuta

2020 ◽  
Vol 34 (S1) ◽  
pp. 1-1
Author(s):  
Sruthi Rajeev ◽  
Manon Defaye ◽  
Adam Shute ◽  
Arthur Wang ◽  
Susan Wang ◽  
...  
2020 ◽  
Vol 3 (Supplement_1) ◽  
pp. 55-56
Author(s):  
S Rajeev ◽  
M Defaye ◽  
A J Shute ◽  
A Wang ◽  
S J Wang ◽  
...  

Abstract Background Tuft cells are a rare chemosensory population of the intestinal epithelium that detect intestinal parasitic nematodes and release IL-25 to mobilize innate lymphoid type 2 cells (ILC2), which then drive a Th2- dominant nematode expulsion response. Immunocompetent mice develop tuft cell hyperplasia in the small intestine during infection with Hymenolepis diminuta, a non-abrasive lumen dwelling small intestinal cestode parasite. Helminth infections are accompanied by alterations in sensory motor functions of the gut as well as the composition of the microbiota. It is poorly understood if tuft cell hyperplasia is regulated by these immunomodulatory influences. Aims To test if mice lacking (1) a functional adaptive immune system, (2) TRPV1+ gut-innervating sensory nerves and (3) a microbiome, display enteric tuft cell hyperplasia following infection with H. diminuta. Methods RAG-1-/- (male and female) and germ-free mice (n=3–6) were infected with 5 cysticercoids of H. diminuta and age matched non-infected mice served as control groups. Male C57BL/6j mice were treated with resiniferatoxin (RTX) to ablate TRPV1 +sensory neurons before infection. Mid-jejunum cryostat or paraffin embedded sections immunostained against doublecortin-like kinase -1 (DCLK-1) were blindly scored for tuft cell enumeration at 5–14 days post-infection. Results Tuft cell hyperplasia (~10-15-fold increase) was observed in the jejunum of wild-type mice at 11 days post infection with H. diminuta, by which time worms are expelled. Infected RAG-1-/- mice develop tuft cell hyperplasia of lesser magnitude than wild-type mice. Germ-free mice displayed tuft cell hyperplasia and kinetics of worm expulsion that were not different from wild-type mice. RTX-treated mice with confirmed loss of TRPV1+ nerve fibers in the gut and their cell soma in the dorsal root and nodose ganglia, had a greater increase (~2-fold) in tuft cell numbers compared to H. diminuta-only mice at 11 days post-infection. Conclusions Knowledge of how the host senses helminths in the gut lumen is central to the host-parasite interaction. Using the H. diminuta-mouse model system we find that tuft cell hyperplasia is largely, but not entirely dependent on adaptive immunity, occurs independent of the gut microbiota, and, intriguingly, TRPV1+ sensory nerves appear to act as a brake on the system, limiting the magnitude of the hyperplasia. Funding Agencies CIHRNSERC, Henry Koopman’s Memorial scholarship


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Zheng Fu ◽  
Joseph W. Dean ◽  
Lifeng Xiong ◽  
Michael W. Dougherty ◽  
Kristen N. Oliff ◽  
...  

AbstractRORγt+ lymphocytes, including interleukin 17 (IL-17)-producing gamma delta T (γδT17) cells, T helper 17 (Th17) cells, and group 3 innate lymphoid cells (ILC3s), are important immune regulators. Compared to Th17 cells and ILC3s, γδT17 cell metabolism and its role in tissue homeostasis remains poorly understood. Here, we report that the tissue milieu shapes splenic and intestinal γδT17 cell gene signatures. Conditional deletion of mitochondrial transcription factor A (Tfam) in RORγt+ lymphocytes significantly affects systemic γδT17 cell maintenance and reduces ILC3s without affecting Th17 cells in the gut. In vivo deletion of Tfam in RORγt+ lymphocytes, especially in γδT17 cells, results in small intestine tissue remodeling and increases small intestine length by enhancing the type 2 immune responses in mice. Moreover, these mice show dysregulation of the small intestine transcriptome and metabolism with less body weight but enhanced anti-helminth immunity. IL-22, a cytokine produced by RORγt+ lymphocytes inhibits IL-13-induced tuft cell differentiation in vitro, and suppresses the tuft cell-type 2 immune circuit and small intestine lengthening in vivo, highlighting its key role in gut tissue remodeling.


Parasitology ◽  
1968 ◽  
Vol 58 (1) ◽  
pp. 227-229 ◽  
Author(s):  
D. W. T. Crompton ◽  
P. J. Whitfield

The cestode, Hymenolepis diminuta, and the acanthocephalan, Moniliformis dubius, were found, by Chandler (1939) and Burlingame & Chandler (1941) respectively, to undergo an anterior migration in the intestine of rats after the establishment of the infection. More recently, these results have been confirmed by Holmes (1961, 1962) who stated that it is not unlikely that the worms select optimum sites along one or more of the many gradients known or postulated to exist along the length of the small intestine.


2020 ◽  
Vol 5 (43) ◽  
pp. eaay3994 ◽  
Author(s):  
Mindy M. Miller ◽  
Preeyam S. Patel ◽  
Katherine Bao ◽  
Thomas Danhorn ◽  
Brian P. O’Connor ◽  
...  

A transitory, interleukin-25 (IL-25)–responsive, group 2 innate lymphoid cell (ILC2) subset induced during type 2 inflammation was recently identified as iILC2s. This study focuses on understanding the significance of this population in relation to tissue-resident nILC2s in the lung and intestine. RNA-sequencing and pathway analysis revealed the AP-1 superfamily transcription factor BATF (basic leucine zipper transcription factor, activating transcription factor–like) as a potential modulator of ILC2 cell fate. Infection of BATF-deficient mice with Nippostrongylus brasiliensis showed a selective defect in IL-25–mediated helminth clearance and a corresponding loss of iILC2s in the lung characterized as IL-17RBhigh, KLRG1high, BATFhigh, and Arg1low. BATF deficiency selectively impaired iILC2s because it had no impact on tissue-resident nILC2 frequency or function. Pulmonary-associated iILC2s migrated to the lung after infection, where they represented an early source of IL-4 and IL-13. Although the composition of ILC2s in the small intestine was distinct from those in the lung, their frequency and IL-13 expression remained dependent on BATF, which was also required for optimal goblet and tuft cell hyperplasia. Findings support IL-25–responsive ILC2s as early sentinels of mucosal barrier integrity.


1970 ◽  
Vol 48 (4) ◽  
pp. 761-769 ◽  
Author(s):  
Christine E. Cannon ◽  
D. F. Mettrick

The changes in the distribution of Hymenolepis diminuta within the rat intestine have been followed over the period 3 to 16 days postinfection using rats, each infected with 10 cysticercoids of H. diminuta, fed ad libitum on Purina Rat Chow.The parameters investigated were distribution of scolex attachment sites in the intestine, and distribution of parasite biomass in the intestine based on strobila length, ex vivo weight distribution, and in vivo weight distribution. Both the scoleces and biomass of 3- and 5-day-old worms are concentrated in the second quarter of the intestine. The mean scolex attachment point for 5-day-old worms was 39% of the total intestinal length behind the pyloric sphincter. Between days 5 and 7 there was a marked anterior migration of the young worms, so that at 7 days the mean scolex attachment site was 15% of the total intestinal length behind the stomach. Over the same period of time the mean in vivo weight distribution moved forward from a point 44% behind the pyloric sphincter to one only 23% along the intestine. After 7 days there was a gradual posteriad spreading of the scolex attachment sites and of the parasite biomass. Hymenolepis diminuta can attach itself anywhere in the anterior 75% of the intestine, including in front of the opening of the bile duct: no worms were found in the small intestine extending back into the caecum.The pattern of migration and the changes in worm distribution in the intestine suggest that H. diminuta selects an appropriate, but changing position, on one or more of the gradients that have been demonstrated or postulated along the length of the small intestine.It is also suggested that the long-term migration during prepatent development is interrelated, but distinct from the daily migrational movements that H. diminuta undergoes within the small intestine.


Parasitology ◽  
1981 ◽  
Vol 83 (3) ◽  
pp. 489-496 ◽  
Author(s):  
Erling Bindseil ◽  
Jørn Andreassen

SUMMARYMice inoculated with 2000 Ascaris suum eggs 7 days before an infection with 2 cysticercoids of Hymenolepis diminuta harboured significantly fewer and/or smaller tapeworms than control mice by day 7 post-infection. When the interval between the infections was increased, the effect on H. diminuta decreased and no effect was found 21 days after the A. suum infection or if the infections were given simultaneously in ńaive or in mice immune to A. suum. Two possible explanations for the rejection and/or stunting of H. diminuta in mice infected 7 days earlier with A. suum are suggested; either a host reaction in the small intestine stimulated by the returning larvae of A. suum after their hepato–pulmonary migration or an indirect effect of decreased food intake of the host caused by this migration. It is concluded that experiments on possible immunodepressive or immunostimulating effects of parasites ought to include studies on living agents and that they should not rely on measurements of immune responses only.


Sign in / Sign up

Export Citation Format

Share Document