Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.704
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 39: 639-665, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33646858

ABSTRACT

Coevolutionary adaptation between humans and helminths has developed a finely tuned balance between host immunity and chronic parasitism due to immunoregulation. Given that these reciprocal forces drive selection, experimental models of helminth infection are ideally suited for discovering how host protective immune responses adapt to the unique tissue niches inhabited by these large metazoan parasites. This review highlights the key discoveries in the immunology of helminth infection made over the last decade, from innate lymphoid cells to the emerging importance of neuroimmune connections. A particular emphasis is placed on the emerging areas within helminth immunology where the most growth is possible, including the advent of genetic manipulation of parasites to study immunology and the use of engineered T cells for therapeutic options. Lastly,we cover the status of human challenge trials with helminths as treatment for autoimmune disease, which taken together, stand to keep the study of parasitic worms at the forefront of immunology for years to come.


Subject(s)
Helminthiasis , Helminths , Parasites , Animals , Host-Parasite Interactions , Humans , Immunity, Innate , Lymphocytes , T-Lymphocytes
2.
Annu Rev Immunol ; 37: 47-72, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30379593

ABSTRACT

Tuft cells-rare solitary chemosensory cells in mucosal epithelia-are undergoing intense scientific scrutiny fueled by recent discovery of unsuspected connections to type 2 immunity. These cells constitute a conduit by which ligands from the external space are sensed via taste-like signaling pathways to generate outputs unique among epithelial cells: the cytokine IL-25, eicosanoids associated with allergic immunity, and the neurotransmitter acetylcholine. The classic type II taste cell transcription factor POU2F3 is lineage defining, suggesting a conceptualization of these cells as widely distributed environmental sensors with effector functions interfacing type 2 immunity and neural circuits. Increasingly refined single-cell analytics have revealed diversity among tuft cells that extends from nasal epithelia and type II taste cells to ex-Aire-expressing medullary thymic cells and small-intestine cells that mediate tissue remodeling in response to colonizing helminths and protists.


Subject(s)
Epithelium/physiology , Helminthiasis/immunology , Helminths/physiology , Octamer Transcription Factors/metabolism , Sensory Receptor Cells/physiology , Th2 Cells/immunology , Animals , Humans , Immune System , Interleukin-17/metabolism , Nervous System , Neuroimmunomodulation , Octamer Transcription Factors/genetics , Signal Transduction , TRPM Cation Channels/metabolism
3.
Annu Rev Immunol ; 35: 53-84, 2017 04 26.
Article in English | MEDLINE | ID: mdl-27912316

ABSTRACT

Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.


Subject(s)
Helminthiasis/immunology , Hypersensitivity/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Disease Models, Animal , Epigenesis, Genetic , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunity, Humoral , Immunologic Memory , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism
4.
Annu Rev Immunol ; 33: 201-25, 2015.
Article in English | MEDLINE | ID: mdl-25533702

ABSTRACT

Helminth parasites are a highly successful group of pathogens that challenge the immune system in a manner distinct from rapidly replicating infectious agents. Of this group, roundworms (nematodes) that dwell in the intestines of humans and other animals are prevalent worldwide. Currently, more than one billion people are infected by at least one species, often for extended periods of time. Thus, host-protective immunity is rarely complete. The reasons for this are complex, but laboratory investigation of tractable model systems in which protective immunity is effective has provided a mechanistic understanding of resistance that is characterized almost universally by a type 2/T helper 2 response. Greater understanding of the mechanisms of susceptibility has also provided the basis for defining host immunoregulation and parasite-evasion strategies, helping place in context the changing patterns of immunological disease observed worldwide.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Host-Pathogen Interactions/immunology , Adaptive Immunity , Animals , Antigens, Helminth/immunology , Disease Resistance , Disease Susceptibility , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/parasitology , Gastrointestinal Microbiome , Humans , Immunity, Innate , Nematoda/immunology , Nematode Infections/immunology , Nematode Infections/microbiology , Nematode Infections/parasitology
5.
Immunity ; 57(5): 935-937, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38749395

ABSTRACT

The intestinal epithelium interacts with immune cells to support tissue homeostasis and coordinate responses against pathogens. In this issue of Immunity, Yang et al. unveil a central role for mast cell-epithelial cell interactions in orchestrating protective type 2 immune responses following intestinal helminth infection.


Subject(s)
Intestinal Mucosa , Mast Cells , Mast Cells/immunology , Animals , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Humans , Homeostasis/immunology , Helminthiasis/immunology , Helminthiasis/parasitology , Epithelial Cells/immunology , Mice
6.
Immunity ; 57(6): 1260-1273.e7, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38744292

ABSTRACT

Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.


Subject(s)
Acetylcholine , Intestinal Mucosa , Animals , Acetylcholine/metabolism , Mice , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Choline O-Acetyltransferase/metabolism , Interleukin-13/metabolism , Interleukin-13/immunology , Mice, Knockout , Mice, Inbred C57BL , Helminthiasis/immunology , Helminthiasis/parasitology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Immunity, Innate , Nematospiroides dubius/immunology , Tuft Cells
7.
Immunity ; 55(4): 623-638.e5, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385697

ABSTRACT

The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.


Subject(s)
Alarmins , Intestinal Mucosa , Acylation , Alarmins/immunology , Anthelmintics/immunology , Biomarkers, Tumor , Cytokines , DNA-Binding Proteins , Helminthiasis/immunology , Humans , Hyperplasia , Inflammation , Interleukin-33 , Intestinal Mucosa/immunology , Mebendazole , N-Acetylglucosaminyltransferases/immunology , Pore Forming Cytotoxic Proteins , STAT6 Transcription Factor/immunology
9.
Nat Immunol ; 16(10): 1051-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322482

ABSTRACT

Type 2 helper T cells (TH2 cells) produce interleukin 13 (IL-13) when stimulated by papain or house dust mite extract (HDM) and induce eosinophilic inflammation. This innate response is dependent on IL-33 but not T cell antigen receptors (TCRs). While type 2 innate lymphoid cells (ILC2 cells) are the dominant innate producers of IL-13 in naive mice, we found here that helminth-infected mice had more TH2 cells compared to uninfected mice, and thes e cells became major mediators of innate type 2 responses. TH2 cells made important contributions to HDM-induced antigen-nonspecific eosinophilic inflammation and protected mice recovering from infection with Ascaris suum against subsequent infection with the phylogenetically distant nematode Nippostrongylus brasiliensis. Our findings reveal a previously unappreciated role for effector TH2 cells during TCR-independent innate-like immune responses.


Subject(s)
Immunity, Innate , Th2 Cells/immunology , Animals , Flow Cytometry , Helminthiasis/immunology , Helminths/immunology , Lung/cytology , Lung/immunology , Lymphocytes/immunology , Mice , Polymerase Chain Reaction
10.
Immunity ; 49(5): 801-818, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30462997

ABSTRACT

Helminths are extraordinarily successful parasites due to their ability to modulate the host immune response. They have evolved a spectrum of immunomodulatory molecules that are now beginning to be defined, heralding a molecular revolution in parasite immunology. These discoveries have the potential both to transform our understanding of parasite adaptation to the host and to develop possible therapies for immune-mediated disease. In this review we will summarize the current state of the art in parasite immunomodulation and discuss perspectives on future areas for research and discovery.


Subject(s)
Helminthiasis/immunology , Helminths/immunology , Host-Parasite Interactions , Immunomodulation , Adaptive Immunity , Animals , Biological Evolution , Dendritic Cells/immunology , Dendritic Cells/metabolism , Helminthiasis/parasitology , Humans , Immunity, Innate , Macrophages/immunology , Macrophages/metabolism
11.
Proc Natl Acad Sci U S A ; 121(25): e2308733121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857387

ABSTRACT

The WHO recommends mass drug administration (MDA) for intestinal worm infections in areas with over 20% infection prevalence. Recent Cochrane meta-analyses endorse treatment of infected individuals but recommend against MDA. We conducted a theory-agnostic random-effects meta-analysis of the effect of multiple-dose MDA and a cost-effectiveness analysis. We estimate significant effects of MDA on child weight (0.15 kg, 95% CI: 0.07, 0.24; P < 0.001), mid-upper arm circumference (0.20 cm, 95% CI: 0.03, 0.37; P = 0.02), and height (0.09 cm, 95% CI: 0.01, 0.16; P = 0.02) when prevalence is over 20% but not on Hb (0.06 g/dL, 95% CI: -0.01, 0.14; P = 0.1). These results suggest that MDA is a cost-effective intervention, particularly in the settings where it is recommended by the WHO.


Subject(s)
Helminthiasis , Intestinal Diseases, Parasitic , Humans , Intestinal Diseases, Parasitic/drug therapy , Intestinal Diseases, Parasitic/epidemiology , Helminthiasis/drug therapy , Helminthiasis/epidemiology , Mass Drug Administration , Anthelmintics/therapeutic use , Anthelmintics/administration & dosage , Public Policy , Cost-Benefit Analysis , Child
12.
Immunity ; 47(6): 1024-1036, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262347

ABSTRACT

Type-2-cell-mediated immune responses play a critical role in mediating both host-resistance and disease-tolerance mechanisms during helminth infections. Recently, type 2 cell responses have emerged as major regulators of tissue repair and metabolic homeostasis even under steady-state conditions. In this review, we consider how studies of helminth infection have contributed toward our expanding cellular and molecular understanding of type-2-cell-mediated immunity, as well as new areas such as the microbiome. By studying how these successful parasites form chronic infections without overt pathology, we are gaining additional insights into allergic and inflammatory diseases, as well as normal physiology.


Subject(s)
Helminthiasis/immunology , Immunity, Cellular , Macrophages/immunology , Nematoda/immunology , Th2 Cells/immunology , Trematoda/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/parasitology , Gene Expression Regulation/immunology , Helminthiasis/genetics , Helminthiasis/parasitology , Homeostasis/immunology , Host-Parasite Interactions/immunology , Humans , Macrophages/parasitology , Mast Cells/immunology , Mast Cells/parasitology , Microbiota/immunology , Th2 Cells/parasitology
13.
Immunity ; 46(5): 863-874.e4, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514691

ABSTRACT

Mast cells are important for eradication of intestinal nematodes; however, their precise mechanisms of action have remained elusive, especially in the early phase of infection. We found that Spi-B-deficient mice had increased numbers of mast cells and rapidly expelled the Heligmosomoides polygyrus (Hp) nematode. This was accompanied by induction of interleukin-13 (IL-13)-producing group 2 innate lymphoid cells (ILC2) and goblet cell hyperplasia. Immediately after Hp infection, mast cells were rapidly activated to produce IL-33 in response to ATP released from apoptotic intestinal epithelial cells. In vivo inhibition of the P2X7 ATP receptor rendered the Spi-B-deficient mice susceptible to Hp, concomitant with elimination of mast cell activation and IL-13-producing ILC2 induction. These results uncover a previously unknown role for mast cells in innate immunity in that activation of mast cells by ATP orchestrates the development of a protective type 2 immune response, in part by producing IL-33, which contributes to ILC2 activation.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Immunity, Innate , Lymphocyte Subsets/immunology , Mast Cells/immunology , Adenosine Triphosphate/metabolism , Animals , Cell Communication , Cell Differentiation , Disease Models, Animal , Disease Resistance/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression , Helminthiasis/genetics , Immunophenotyping , Interleukin-33/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Male , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Knockout , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Immunol ; 212(4): 632-644, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38180236

ABSTRACT

Distinct subsets of T lymphocytes express CX3CR1 under inflammatory conditions, but little is known about CX3CR1+CD4+ T cells during type 2 inflammation in helminth infections. In this study, we used a fate-mapping mouse model to characterize CX3CR1+CD4+ T cells during both acute Nippostrongylus brasiliensis and chronic Schistosoma mansoni murine models of helminth infections, revealing CX3CR1+CD4+ T cells to be an activated tissue-homing subset with varying capacity for cytokine production. Tracking these cells over time revealed that maintenance of CX3CR1 itself along with a TH2 phenotype conferred a survival advantage in the inflamed tissue. Single-cell RNA sequencing analysis of fate-mapped CX3CR1+CD4+ T cells from both the peripheral tissue and the spleen revealed a considerable level of diversity and identified a distinct population of BCL6+TCF-1+PD1+CD4+ T cells in the spleen during helminth infections. Conditional deletion of BCL6 in CX3CR1+ cells resulted in fewer CX3CR1+CD4+ T cells during infection, indicating a role in sustaining CD4+ T cell responses to helminth infections. Overall, our studies revealed the behavior and heterogeneity of CX3CR1+CD4+ T cells during type 2 inflammation in helminth infections and identified BCL6 to be important in their maintenance.


Subject(s)
CD4-Positive T-Lymphocytes , Helminthiasis , Schistosoma mansoni , Animals , Mice , CD4-Positive T-Lymphocytes/metabolism , Helminthiasis/immunology , Inflammation/metabolism , Schistosoma mansoni/physiology
15.
Clin Microbiol Rev ; 37(1): e0009823, 2024 03 14.
Article in English | MEDLINE | ID: mdl-38319102

ABSTRACT

Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.


Subject(s)
Coinfection , Helminthiasis , Schistosomiasis , Humans , Coinfection/epidemiology , Coinfection/parasitology , Schistosomiasis/complications , Schistosomiasis/epidemiology , Schistosomiasis/parasitology , Africa , Soil/parasitology , Prevalence , Helminthiasis/complications , Helminthiasis/epidemiology , Helminthiasis/parasitology
16.
Nat Immunol ; 14(11): 1118-1126, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24145791

ABSTRACT

Helminth infections are ubiquitous worldwide and can trigger potent immune responses that differ from and potentially antagonize host protective responses to microbial pathogens. In this Review we focus on the three main killers in infectious disease-AIDS, tuberculosis and malaria-and critically assesses whether helminths adversely influence host control of these diseases. We also discuss emerging concepts for how M2 macrophages and helminth-modulated dendritic cells can potentially influence the protective immune response to concurrent infections. Finally, we present evidence advocating for more efforts to determine how and to what extent helminths interfere with the successful control of specific concurrent coinfections.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Dendritic Cells/immunology , Helminthiasis/immunology , Macrophages/immunology , Malaria/immunology , Tuberculosis/immunology , Acquired Immunodeficiency Syndrome/epidemiology , Acquired Immunodeficiency Syndrome/virology , Africa/epidemiology , Animals , Asia/epidemiology , Coinfection , Dendritic Cells/microbiology , Dendritic Cells/parasitology , Dendritic Cells/virology , Helminthiasis/epidemiology , Helminthiasis/parasitology , Helminths/immunology , Host-Parasite Interactions , Humans , Latin America/epidemiology , Macrophages/microbiology , Macrophages/parasitology , Macrophages/virology , Malaria/epidemiology , Malaria/parasitology , Tuberculosis/epidemiology , Tuberculosis/microbiology
17.
J Immunol ; 211(3): 389-402, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37272847

ABSTRACT

The impact of endemic parasitic infection on vaccine efficacy is an important consideration for vaccine development and deployment. We have examined whether intestinal infection with the natural murine helminth Heligmosomoides polygyrus bakeri alters Ag-specific Ab and cellular immune responses to oral and parenteral vaccination in mice. Oral vaccination of mice with a clinically relevant, live, attenuated, recombinant Salmonella vaccine expressing chicken egg OVA (Salmonella-OVA) induced the accumulation of activated, OVA-specific T effector cells rather than OVA-specific regulatory T cells in the GALT. Intestinal helminth infection significantly reduced Th1-skewed Ab responses to oral vaccination with Salmonella-OVA. Activated, adoptively transferred, OVA-specific CD4+ T cells accumulated in draining mesenteric lymph nodes of vaccinated mice, regardless of their helminth infection status. However, helminth infection increased the frequencies of adoptively transferred OVA-specific CD4+ T cells producing IL-4 and IL-10 in the mesenteric lymph node. Ab responses to the oral Salmonella-OVA vaccine were reduced in helminth-free mice adoptively transferred with OVA-specific CD4+ T cells harvested from mice with intestinal helminth infection. Intestinal helminth infection also significantly reduced Th2-skewed Ab responses to parenteral vaccination with OVA adsorbed to alum. These findings suggest that vaccine-specific CD4+ T cells induced in the context of helminth infection retain durable immunomodulatory properties and may promote blunted Ab responses to vaccination. They also underscore the potential need to treat parasitic infection before mass vaccination campaigns in helminth-endemic areas.


Subject(s)
Helminthiasis , Intestinal Diseases, Parasitic , Mice , Animals , Vaccine Efficacy , CD4-Positive T-Lymphocytes , Vaccines, Synthetic , Ovalbumin , Mice, Inbred BALB C
18.
J Immunol ; 210(3): 297-309, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36524995

ABSTRACT

CD8 virtual memory T (TVM) cells are Ag-naive CD8 T cells that have undergone partial differentiation in response to common γ-chain cytokines, particularly IL-15 and IL-4. TVM cells from young individuals are highly proliferative in response to TCR and cytokine stimulation but, with age, they lose TCR-mediated proliferative capacity and exhibit hallmarks of senescence. Helminth infection can drive an increase in TVM cells, which is associated with improved pathogen clearance during subsequent infectious challenge in young mice. Given the cytokine-dependent profile of TVM cells and their age-associated dysfunction, we traced proliferative and functional changes in TVM cells, compared with true naive CD8 T cells, after helminth infection of young and aged C57BL/6 mice. We show that IL-15 is essential for the helminth-induced increase in TVM cells, which is driven only by proliferation of existing TVM cells, with negligible contribution from true naive cell differentiation. Additionally, TVM cells showed the greatest proliferation in response to helminth infection and IL-15 compared with other CD8 T cells. Furthermore, TVM cells from aged mice did not undergo expansion after helminth infection due to both TVM cell-intrinsic and -extrinsic changes associated with aging.


Subject(s)
Helminthiasis , Interleukin-15 , Animals , Mice , Aging/immunology , CD8-Positive T-Lymphocytes/parasitology , Cytokines , Helminthiasis/immunology , Helminthiasis/metabolism , Helminths/pathogenicity , Immunologic Memory , Interleukin-15/metabolism , Mice, Inbred C57BL , Receptors, Antigen, T-Cell
19.
Semin Immunol ; 53: 101532, 2021 03.
Article in English | MEDLINE | ID: mdl-34823996

ABSTRACT

IL-33 is an alarmin cytokine which has been implicated in allergy, fibrosis, inflammation, tumorigenesis, metabolism, and homeostasis. However, amongst its strongest roles are in helminth infections, where IL-33 usually (but not always) is central to induction of an effective anti-parasitic immune response. In this review, we will summarise the literature around this fascinating cytokine, its activity on immune and non-immune cells, the unique (and sometimes counterintuitive) responses it induces, and how it can coordinate the immune response during infections by parasitic helminths. Finally, we will summarise some of the ways that parasites have developed to modulate the IL-33 pathway for their own benefit.


Subject(s)
Helminthiasis , Helminths , Hypersensitivity , Interleukin-33/metabolism , Animals , Cytokines/metabolism , Helminthiasis/parasitology , Helminths/metabolism , Humans
20.
Clin Infect Dis ; 78(Supplement_2): S146-S152, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662703

ABSTRACT

Globally, there are over 1 billion people infected with soil-transmitted helminths (STHs), mostly living in marginalized settings with inadequate sanitation in sub-Saharan Africa and Southeast Asia. The World Health Organization recommends an integrated approach to STH morbidity control through improved access to sanitation and hygiene education and the delivery of preventive chemotherapy (PC) to school-age children delivered through schools. Progress of STH control programs is currently estimated using a baseline (pre-PC) school-based prevalence survey and then monitored using periodical school-based prevalence surveys, known as Impact Assessment Surveys (IAS). We investigated whether integrating geostatistical methods with a Markov model or a mechanistic transmission model for projecting prevalence forward in time from baseline can improve IAS design strategies. To do this, we applied these 2 methods to prevalence data collected in Kenya, before evaluating and comparing their performance in accurately informing optimal survey design for a range of IAS sampling designs. We found that, although both approaches performed well, the mechanistic method more accurately projected prevalence over time and provided more accurate information for guiding survey design. Both methods performed less well in areas with persistent STH hotspots where prevalence did not decrease despite multiple rounds of PC. Our findings show that these methods can be useful tools for more efficient and accurate targeting of PC. The general framework built in this paper can also be used for projecting prevalence and informing survey design for other neglected tropical diseases.


Subject(s)
Helminthiasis , Markov Chains , Soil , Humans , Helminthiasis/epidemiology , Helminthiasis/transmission , Prevalence , Kenya/epidemiology , Soil/parasitology , Child , Helminths/isolation & purification , Animals , Models, Statistical , Adolescent , Schools
SELECTION OF CITATIONS
SEARCH DETAIL