Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Front Immunol ; 12: 728848, 2021.
Article in English | MEDLINE | ID: mdl-34557194

ABSTRACT

Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.


Subject(s)
Adaptive Immunity , Immune System/parasitology , Immunity, Innate , Leishmania braziliensis/pathogenicity , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Visceral/parasitology , Phagocytosis , Phagosomes/parasitology , Animals , Chronic Disease , Host-Parasite Interactions , Humans , Immune System/immunology , Immune System/metabolism , Leishmania braziliensis/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/metabolism , Leishmaniasis, Mucocutaneous/immunology , Leishmaniasis, Mucocutaneous/metabolism , Leishmaniasis, Mucocutaneous/parasitology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/metabolism , Phagosomes/immunology , Phagosomes/metabolism
3.
Front Immunol ; 12: 667787, 2021.
Article in English | MEDLINE | ID: mdl-34054837

ABSTRACT

Freshwater crayfish immunity has received great attention due to the need for urgent conservation. This concern has increased the understanding of the cellular and humoral defense systems, although the regulatory mechanisms involved in these processes need updating. There are, however, aspects of the immune response that require clarification and integration. The particular issues addressed in this review include an overall description of the oomycete Aphanomyces astaci, the causative agent of the pandemic plague disease, which affects freshwater crayfish, and an overview of crustaceans' immunity with a focus on freshwater crayfish. It includes a classification system of hemocyte sub-types, the molecular factors involved in hematopoiesis and the differential role of the hemocyte subpopulations in cell-mediated responses, including hemocyte infiltration, inflammation, encapsulation and the link with the extracellular trap cell death pathway (ETosis). In addition, other topics discussed include the identity and functions of hyaline cells, the generation of neoplasia, and the emerging topic of the role of sessile hemocytes in peripheral immunity. Finally, attention is paid to the molecular execution of the immune response, from recognition by the pattern recognition receptors (PRRs), the role of the signaling network in propagating and maintaining the immune signals, to the effector elements such as the putative function of the Down syndrome adhesion molecules (Dscam) in innate immune memory.


Subject(s)
Aphanomyces/pathogenicity , Astacoidea/parasitology , Immune System/parasitology , Immunity, Innate , Infections/veterinary , Animals , Aphanomyces/immunology , Astacoidea/immunology , Astacoidea/metabolism , Fresh Water , Hemocytes/immunology , Hemocytes/metabolism , Hemocytes/parasitology , Host-Parasite Interactions , Immune System/immunology , Immune System/metabolism , Infections/immunology , Infections/metabolism , Infections/parasitology , Receptors, Pattern Recognition/metabolism , Signal Transduction
4.
Cell Mol Life Sci ; 78(10): 4545-4561, 2021 May.
Article in English | MEDLINE | ID: mdl-33713154

ABSTRACT

Malaria is a vector-borne parasitic disease with a vast impact on human history, and according to the World Health Organisation, Plasmodium parasites still infect over 200 million people per year. Plasmodium falciparum, the deadliest parasite species, has a remarkable ability to undermine the host immune system and cause life-threatening disease during blood infection. The parasite's host cells, red blood cells (RBCs), generally maintain an asymmetric distribution of phospholipids in the two leaflets of the plasma membrane bilayer. Alterations to this asymmetry, particularly the exposure of phosphatidylserine (PS) in the outer leaflet, can be recognised by phagocytes. Because of the importance of innate immune defence numerous studies have investigated PS exposure in RBCs infected with P. falciparum, but have reached different conclusions. Here we review recent advancements in our understanding of the molecular mechanisms which regulate asymmetry in RBCs, and whether infection with the P. falciparum parasite results in changes to PS exposure. On the balance of evidence, it is likely that membrane asymmetry is disrupted in parasitised RBCs, though some methodological issues need addressing. We discuss the potential causes and consequences of altered asymmetry in parasitised RBCs, particularly for in vivo interactions with the immune system, and the role of host-parasite co-evolution. We also examine the potential asymmetric state of parasite membranes and summarise current knowledge on the parasite proteins, which could regulate asymmetry in these membranes. Finally, we highlight unresolved questions at this time and the need for interdisciplinary approaches to uncover the machinery which enables P. falciparum parasites to hide in mature erythrocytes.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/parasitology , Erythrocytes/metabolism , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Phospholipids/metabolism , Plasmodium falciparum/pathogenicity , Animals , Erythrocytes/parasitology , Host-Parasite Interactions/physiology , Humans , Immune System/metabolism , Immune System/parasitology
5.
Yale J Biol Med ; 92(2): 225-231, 2019 06.
Article in English | MEDLINE | ID: mdl-31249483

ABSTRACT

From bacteria to mammals, nearly all organisms have adapted their physiology and behavior to a daily rhythm. These circadian (daily) rhythms influence virtually all aspects of physiological architecture (i.e., from gene expression to organismal behavior). Therefore, it is not surprising that several features of the immune response are regulated in a time-of-day dependent manner. The field of chrono-immunology has expanded tremendously over the past decade. In this abridged review, we present studies from the past five years that have revealed new parameters of the immune system that demonstrate daily variations in the control of pathogens and response to microbial components. These studies analyzed how the disruption of circadian rhythms impairs immune function, how microbial components alter the circadian clock, and how immune responses demonstrate daily variations in human subjects. Further elucidating the intricate connections between the circadian clock and the immune system will hopefully provide opportunities for chrono-immunotherapy in disease treatment and prevention.


Subject(s)
Circadian Clocks/immunology , Circadian Rhythm/immunology , Immune System/immunology , Infections/immunology , Mammals/immunology , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/immunology , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Gene Expression Regulation/immunology , Humans , Immune System/microbiology , Immune System/parasitology , Infections/microbiology , Infections/parasitology , Mammals/microbiology , Mammals/parasitology
6.
J Helminthol ; 94: e53, 2019 May 16.
Article in English | MEDLINE | ID: mdl-31092301

ABSTRACT

Neglected tropical diseases (NTDs) share certain traits: they are parasitic infections, prevailing in tropical environments and affecting marginalized sectors of the population. Six NTDs - ascariasis, cysticercosis, echinococcosis, hookworm infection, onchocerciasis and trichuriasis - all of them endemic in Latin America and the Caribbean (LAC), are analysed in this work. This review aims to discuss key information on the function of excretory/secretory (E/S) proteins from these parasites in their infectivity, pathogeny and diagnosis. The modulation of the host immune system to favour the permanence and survival of the parasite is also discussed. An updated knowledge on the function of E/S molecules in endemic parasitoses in LAC may lead to new approaches for the clinical management and diagnosis of these diseases. In turn, this could allow us to optimize their treatment and make it more affordable - a relevant goal given the economic constraints that the region is facing.


Subject(s)
Endemic Diseases , Helminth Proteins/physiology , Helminthiasis/epidemiology , Immune System/parasitology , Neglected Diseases/parasitology , Animals , Caribbean Region/epidemiology , Disease Management , Helminthiasis/immunology , Helminthiasis/parasitology , Host-Parasite Interactions , Humans , Latin America/epidemiology , Neglected Diseases/epidemiology , Neglected Diseases/immunology , Tropical Medicine
7.
BMC Genomics ; 19(1): 932, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30547741

ABSTRACT

BACKGROUND: Regulatory circuits of infection in the emerging experimental model system, water flea Daphnia and their microparasites, remain largely unknown. Here we provide the first molecular insights into the response of Daphnia galeata to its highly virulent and common parasite Caullerya mesnili, an ichthyosporean that infects the gut epithelium. We generated a transcriptomic dataset using RNAseq from parasite-exposed (vs. control) Daphnia, at two time points (4 and 48 h) after parasite exposure. RESULTS: We found a down-regulation of metabolism and immunity-related genes, at 48 h (but not 4 h) after parasite exposure. These genes are involved in lipid metabolism and fatty acid biosynthesis, as well as microbe recognition (e.g. c-type lectins) and pathogen attack (e.g. gut chitin). CONCLUSIONS: General metabolic suppression implies host energy shift from reproduction to survival, which is in agreement with the known drastic reduction in Daphnia fecundity after Caullerya infection. The down-regulation of gut chitin indicates a possible interaction between the peritrophic matrix and the evading host immune system. Our study provides the first description of host transcriptional responses in this very promising host-parasite experimental system.


Subject(s)
Daphnia/genetics , Immune System/metabolism , Intestines/parasitology , Lipid Metabolism/genetics , Mesomycetozoea/physiology , Animals , Daphnia/metabolism , Down-Regulation , Fatty Acid Synthases/genetics , Host-Parasite Interactions , Immune System/parasitology , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Sequence Analysis, RNA , Transcriptome
8.
PLoS Negl Trop Dis ; 12(5): e0006439, 2018 05.
Article in English | MEDLINE | ID: mdl-29771921

ABSTRACT

Mosquito saliva is a very complex concoction of >100 proteins, many of which have unknown functions. The effects of mosquito saliva proteins injected into our skin during blood feeding have been studied mainly in mouse models of injection or biting, with many of these systems producing results that may not be relevant to human disease. Here, we describe the numerous effects that mosquito bites have on human immune cells in mice engrafted with human hematopoietic stem cells. We used flow cytometry and multiplex cytokine bead array assays, with detailed statistical analyses, to detect small but significant variations in immune cell functions after 4 mosquitoes fed on humanized mice footpads. After preliminary analyses, at different early times after biting, we focused on assessing innate immune and subsequent cellular responses at 6 hours, 24 hours and 7 days after mosquito bites. We detected both Th1 and Th2 human immune responses, and delayed effects on cytokine levels in the blood, and immune cell compositions in the skin and bone marrow, up to 7 days post-bites. These are the first measurements of this kind, with human immune responses in whole animals, bitten by living mosquitoes, versus previous studies using incomplete mouse models and salivary gland extracts or needle injected saliva. The results have major implications for the study of hematophagous insect saliva, its effects on the human immune system, with or without pathogen transmission, and the possibility of determining which of these proteins to target for vaccination, in attempts to block transmission of numerous tropical diseases.


Subject(s)
Aedes/immunology , Immune System/immunology , Insect Bites and Stings/immunology , Saliva/immunology , Aedes/physiology , Animals , Cytokines/genetics , Cytokines/immunology , Female , Humans , Immune System/parasitology , Insect Bites and Stings/genetics , Insect Bites and Stings/parasitology , Insect Proteins/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/immunology
9.
HLA ; 91(4): 255-270, 2018 04.
Article in English | MEDLINE | ID: mdl-29368453

ABSTRACT

Little attention has been devoted to the role of HLA-G gene and molecule on parasitic disorders, and the available studies have focused on malaria, African and American trypanosomiasis, leishmaniosis, toxoplasmosis and echinococcosis. After reporting a brief description regarding the role of the cells of innate and adaptive immune system against parasites, we reviewed the major features of the HLA-G gene and molecule and the role of HLA-G on the major cells of immune system. Increased levels of soluble HLA-G (sHLA-G) have been observed in patients presenting toxoplasmosis and in the active phase of echinococcosis. In addition, increased sHLA-G has also been associated with increased susceptibility to malaria and increased susceptibility to develop human African trypanosomiasis (HAT). In contrast, decreased membrane-bound HLA-G has been reported in placenta of patients infected with Plasmodium falciparum and in heart and colon of patients presenting Chagas disease. The 3' untranslated region of the HLA-G gene has been the main focus of studies on malaria, HAT and Chagas disease, exhibiting distinct patterns of associations. Considering that HLA-G is an immune checkpoint molecule, inhibiting the activity of several cells of the immune system, the excessive neoexpression and the increased sHLA-G levels together with the decreased constitutive tissue expression of membrane-bound HLA-G may be detrimental to the host infected with parasite agents.


Subject(s)
HLA-G Antigens/metabolism , Parasitic Diseases/immunology , HLA-G Antigens/genetics , Humans , Immune System/parasitology , Immunity
10.
Semin Immunopathol ; 38(2): 249-60, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26678994

ABSTRACT

The Leishmania granuloma shares some, though not all, properties with that formed following mycobacterial infection. As a simplified, noncaseating granuloma composed of relatively few and largely mononuclear cell populations, it provides a tractable model system to investigate intra-granuloma cellular dynamics, immune regulation, and antimicrobial resistance. Here, the occurrence of granulomatous pathology across the spectrum of leishmaniasis, in humans and animal reservoir hosts, is first described. However, this review focuses on the process of hepatic granuloma formation as studied in rodent models of visceral leishmaniasis, starting from the initial infection of Kupffer cells to the involution of the granuloma after pathogen clearance. It describes how the application of intravital imaging and the use of computational modeling have changed some of our thoughts on granuloma function, and illustrates how host-directed therapies have been used to manipulate granuloma form and function for therapeutic benefit. Where appropriate, lessons that may be equally applicable across the spectrum of granulomatous diseases are highlighted.


Subject(s)
Granuloma/etiology , Granuloma/immunology , Leishmania/physiology , Leishmaniasis/immunology , Leishmaniasis/parasitology , Animals , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Chemokines/metabolism , Cytokines/metabolism , Granuloma/pathology , Humans , Immune System/immunology , Immune System/metabolism , Immune System/parasitology , Immune System/pathology , Leishmaniasis/complications , Liver/immunology , Liver/metabolism , Liver/parasitology , Liver/pathology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoid Tissue/parasitology , Lymphoid Tissue/pathology , Mice , Mononuclear Phagocyte System/immunology , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/parasitology , Mononuclear Phagocyte System/pathology
11.
Parasite Immunol ; 37(3): 150-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25573476

ABSTRACT

Upon infection of humans and animals with Toxoplasma gondii, the parasites persist as intraneuronal cysts that are controlled, but not eliminated by the immune system. In particular, intracerebral T cells are crucial in the control of T. gondii infection and are supported by essential functions from other leukocyte populations. Additionally, brain-resident cells including astrocytes, microglia and neurons contribute to the intracerebral immune response by the production of cytokines, chemokines and expression of immunoregulatory cell surface molecules, such as major histocompatibility (MHC) antigens. However, the in vivo behaviour of these individual cell populations, specifically their interaction during cerebral toxoplasmosis, remains to be elucidated. We discuss here what is known about the function of T cells, recruited myeloid cells and brain-resident cells, with particular emphasis on the potential cross-regulation of these cell populations, in governing cerebral toxoplasmosis.


Subject(s)
Cytokines/biosynthesis , Immune System/immunology , Toxoplasma/physiology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Cerebral/parasitology , Animals , Astrocytes/immunology , Astrocytes/parasitology , Brain/immunology , Brain/parasitology , Chemokines/biosynthesis , Humans , Immune System/parasitology , Microglia/immunology , Microglia/parasitology , Neurons/immunology , Neurons/parasitology , T-Lymphocytes/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Cerebral/immunology
12.
PLoS One ; 9(7): e102821, 2014.
Article in English | MEDLINE | ID: mdl-25054623

ABSTRACT

We introduce and analyze a within-host dynamical model of the coevolution between rapidly mutating pathogens and the adaptive immune response. Pathogen mutation and a homeostatic constraint on lymphocytes both play a role in allowing the development of chronic infection, rather than quick pathogen clearance. The dynamics of these chronic infections display emergent structure, including branching patterns corresponding to asexual pathogen speciation, which is fundamentally driven by the coevolutionary interaction. Over time, continued branching creates an increasingly fragile immune system, and leads to the eventual catastrophic loss of immune control.


Subject(s)
Biological Evolution , Immune System/immunology , Infections/immunology , T-Lymphocytes/immunology , Adaptive Immunity/immunology , Algorithms , Host-Pathogen Interactions/immunology , Humans , Immune System/microbiology , Immune System/parasitology , Infections/microbiology , Infections/parasitology , Models, Immunological , T-Lymphocytes/microbiology , T-Lymphocytes/parasitology
13.
Vet Parasitol ; 206(1-2): 38-42, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24594213

ABSTRACT

The macrocyclic lactones are the only anthelmintics used to prevent heartworm disease, but it is very difficult to reproduce their in vivo efficacy against Dirofilaria immitis larvae in experiments in vitro. These assays typically measure motility, suggesting that paralysis is not the mode of action of the macrocyclic lactones against D. immitis. We isolated peripheral blood mononuclear cells (PBMC) and neutrophils from uninfected dogs and measured their adherence to D. immitis microfilariae in the presence of varying concentrations of ivermectin. We found that adherence of PBMC to the microfilariae was increased in the presence of ivermectin concentrations ≥100 nM and adherence of neutrophils was increased in drug concentrations ≥10 nM. Up to 50% of microfilariae had adherent PBMC in the presence of the drug, and binding was maximal after 40 h incubation. Neutrophil adherence was maximal after 16 h, with approximately 20% of the microfilariae having at least one cell adhered to them. Adherent neutrophils showed morphological evidence of activation. These results are consistent with a model in which the macrocyclic lactones interfere with the parasites ability to evade the host's innate immune system.


Subject(s)
Dirofilaria immitis , Host-Parasite Interactions/drug effects , Ivermectin/pharmacology , Leukocytes, Mononuclear/parasitology , Microfilariae/metabolism , Neutrophils , Animals , Antiparasitic Agents/pharmacology , Dirofilaria immitis/drug effects , Dirofilaria immitis/metabolism , Dogs , Immune System/drug effects , Immune System/parasitology , Leukocytes, Mononuclear/drug effects , Neutrophils/drug effects , Neutrophils/parasitology
14.
J Invest Dermatol ; 134(5): 1211-1219, 2014 May.
Article in English | MEDLINE | ID: mdl-24552683

ABSTRACT

The skin is a critical barrier between hosts and pathogens in arthropod-borne diseases. It harbors many resident cells and specific immune cells to arrest or limit infections by secreting inflammatory molecules or by directly killing pathogens. However, some pathogens are able to use specific skin cells and arthropod saliva for their initial development, to hide from the host immune system, and to establish persistent infection in the vertebrate host. A better understanding of the initial mechanisms taking place in the skin should allow the development of new strategies to fight these vector-borne pathogens that are spread worldwide and are of major medical importance.


Subject(s)
Arthropod Vectors , Arthropods/immunology , Immune System , Infections/transmission , Skin/immunology , Animals , Arthropod Vectors/microbiology , Arthropod Vectors/parasitology , Arthropod Vectors/virology , Humans , Immune System/microbiology , Immune System/parasitology , Immune System/virology , Infections/immunology
15.
Trends Parasitol ; 29(12): 612-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24176554

ABSTRACT

Malaria is a major cause of morbidity and mortality. Immunity is acquired but is suboptimal, being slow to develop and incomplete. An inadequate understanding of natural immunity, host-parasite interactions, and a lack of reliable immune correlates of protection that could predict vaccine efficacy in the field have hindered development of a vaccine. With data from Phase III trials indicating that the leading malaria vaccine candidate, RTS,S, has limited efficacy, it is necessary to reconsider approaches to the development of a vaccine capable of inducing long-lived protection.


Subject(s)
Host-Parasite Interactions/immunology , Immune System/immunology , Immune System/parasitology , Malaria Vaccines/immunology , Malaria/immunology , Animals , Humans , Plasmodium/immunology
16.
PLoS One ; 8(3): e59968, 2013.
Article in English | MEDLINE | ID: mdl-23555851

ABSTRACT

A mathematical model which predicts the intraerythrocytic stages of Plasmodium falciparum infection was developed using data from malaria-infected mice. Variables selected accounted for levels of healthy red blood cells, merozoite (Plasmodium asexual phase) infected red blood cells, gametocyte (Plasmodium sexual phase) infected red blood cells and a phenomenological variable which accounts for the mean activity of the immune system of the host. The model built was able to reproduce the behavior of three different scenarios of malaria. It predicts the later dynamics of malaria-infected humans well after the first peak of parasitemia, the qualitative response of malaria-infected monkeys to vaccination and the changes observed in malaria-infected mice when they are treated with antimalarial drugs. The mathematical model was used to identify new targets to be focused on drug design. Optimization methodologies were applied to identify five targets for minimizing the parasite load; four of the targets thus identified have never before been taken into account in drug design. The potential targets include: 1) increasing the death rate of the gametocytes, 2) decreasing the invasion rate of the red blood cells by the merozoites, 3) increasing the transformation of merozoites into gametocytes, 4) decreasing the activation of the immune system by the gametocytes, and finally 5) a combination of the previous target with decreasing the recycling rate of the red blood cells. The first target is already used in current therapies, whereas the remainders are proposals for potential new targets. Furthermore, the combined target (the simultaneous decrease of the activation of IS by gRBC and the decrease of the influence of IS on the recycling of hRBC) is interesting, since this combination does not affect the parasite directly. Thus, it is not expected to generate selective pressure on the parasites, which means that it would not produce resistance in Plasmodium.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Animals , Drug Design , Drug Discovery , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , Immune System/parasitology , Malaria, Falciparum/physiopathology , Merozoites/physiology , Mice , Models, Theoretical , Parasitemia/drug therapy
17.
Clin Rev Allergy Immunol ; 45(2): 211-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23325330

ABSTRACT

The incidence of autoimmune diseases is increasing in Western countries, possibly due to the improved sanitary conditions and reduced exposure to infections in childhood (the hygiene hypothesis). There is an ongoing debate whether infection prevents or precipitates autoimmune diseases. Various helminths species used in several animal models were shown to limit inflammatory activity in a variety of diseases including inflammatory bowel disease, multiple sclerosis, type 1 diabetes, rheumatoid arthritis, and systemic lupus erythematosus. At present the scientific data is based mostly on experimental animal models; however, there is an increasing body of evidence in a number of clinical trials being conducted. Herein we review several clinical trials evaluating the anti-inflammatory effects of helminths and assessing their association with different autoimmune diseases, including inflammatory bowel disease, multiple sclerosis, and autoimmune liver diseases. We also describe the common pathways by which helminths induce immune modulation and the key changes observed in the host immune system following exposure to helminths. These common pathways include the inhibition of IFN-γ and IL-17 production, promotion of IL-4, IL-10 and TGF-ß release, induction of CD4(+) T cell FoxP3(+) expression, and generation of regulatory macrophages, dendritic cells, and B cells. Helminths products are becoming significant candidates for anti-inflammatory agents in this context. However, further research is needed for synthetic analogues of helminths' potent products that mimic the parasite-mediated immunomodulation effect.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antigens, Helminth/therapeutic use , Autoimmune Diseases/therapy , Helminths/immunology , Therapy with Helminths/methods , Animals , Antigens, Helminth/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/parasitology , Clinical Trials as Topic , Disease Models, Animal , Humans , Immune System/parasitology , Immunomodulation , Ovum/immunology , Therapy with Helminths/trends
18.
J Exp Biol ; 216(Pt 1): 3-10, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225861

ABSTRACT

For millions of years, parasites have altered the behaviour of their hosts. Parasites can affect host behaviour by: (1) interfering with the host's normal immune-neural communication, (2) secreting substances that directly alter neuronal activity via non-genomic mechanisms and (3) inducing genomic- and/or proteomic-based changes in the brain of the host. Changes in host behaviour are often restricted to particular behaviours, with many other behaviours remaining unaffected. Neuroscientists can produce this degree of selectivity by targeting specific brain areas. Parasites, however, do not selectively attack discrete brain areas. Parasites typically induce a variety of effects in several parts of the brain. Parasitic manipulation of host behaviour evolved within the context of the manipulation of other host physiological systems (especially the immune system) that was required for a parasite's survival. This starting point, coupled with the fortuitous nature of evolutionary innovation and evolutionary pressures to minimize the costs of parasitic manipulation, likely contributed to the complex and indirect nature of the mechanisms involved in host behavioural control. Because parasites and neuroscientists use different tactics to control behaviour, studying the methods used by parasites can provide novel insights into how nervous systems generate and regulate behaviour. Studying how parasites influence host behaviour will also help us integrate genomic, proteomic and neurophysiological perspectives on behaviour.


Subject(s)
Brain/parasitology , Host-Parasite Interactions , Parasites/physiology , Animals , Behavior , Behavior, Animal , Biological Evolution , Genomics , Humans , Immune System/parasitology , Neuroimmunomodulation , Neurosciences
19.
J Exp Biol ; 216(Pt 1): 56-66, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225868

ABSTRACT

Parasites affect host behavior in several ways. They can alter activity, microhabitats or both. For trophically transmitted parasites (the focus of our study), decreased activity might impair the ability of hosts to respond to final-host predators, and increased activity and altered microhabitat choice might increase contact rates between hosts and final-host predators. In an analysis of trophically transmitted parasites, more parasite groups altered activity than altered microhabitat choice. Parasites that infected vertebrates were more likely to impair the host's reaction to predators, whereas parasites that infected invertebrates were more likely to increase the host's contact with predators. The site of infection might affect how parasites manipulate their hosts. For instance, parasites in the central nervous system seem particularly suited to manipulating host behavior. Manipulative parasites commonly occupy the body cavity, muscles and central nervous systems of their hosts. Acanthocephalans in the data set differed from other taxa in that they occurred exclusively in the body cavity of invertebrates. In addition, they were more likely to alter microhabitat choice than activity. Parasites in the body cavity (across parasite types) were more likely to be associated with increased host contact with predators. Parasites can manipulate the host through energetic drain, but most parasites use more sophisticated means. For instance, parasites target four physiological systems that shape behavior in both invertebrates and vertebrates: neural, endocrine, neuromodulatory and immunomodulatory. The interconnections between these systems make it difficult to isolate specific mechanisms of host behavioral manipulation.


Subject(s)
Host-Parasite Interactions , Parasites/physiology , Animals , Behavior , Behavior, Animal , Humans , Immune System/parasitology , Nervous System/parasitology , Parasites/classification
20.
Mini Rev Med Chem ; 12(11): 1071-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22924485

ABSTRACT

During the helminth infections, the immune system tends to be modulated by host's sex hormones. Actually, many studies show the reciprocal relationship between sex steroids, the immune system and the elimination or establishment of helminth parasites. Is well known that innate immune response determines the type of adaptive immune response, so the effects in the innate immune response by hormones may affect subsequent adaptive immunity. The sex steroids as estrogens, progesterone and testosterone regulate growth, differentiation, survival and function of many cell types that could be involved in process like homeostasis and immunity, but also have a direct effect on the helminthes, that may probably be mediated by specific receptors on these parasites. Sex steroids, parasites and immunity are closely connected, and their interconnection is involved in the maintenance of elimination or establishment of helminthes in an immunocompetent host. For that reason, understanding the action's mechanisms of sex steroids on immune cells and its direct effect on helminth parasites is important for further progress in the development of novel therapies for chronic helminth diseases associated to immune dysregulation. In this review, we will describe the effects of sex steroids on the immune response during helminth infections as well as the direct effect in these parasites, and the possible implications of these effects on the incidence of several helminth infections.


Subject(s)
Gonadal Steroid Hormones/immunology , Helminthiasis/immunology , Helminths/physiology , Host-Parasite Interactions , Immunity, Innate , Animals , Helminthiasis/parasitology , Helminths/immunology , Humans , Immune System/immunology , Immune System/parasitology , Immunity
SELECTION OF CITATIONS
SEARCH DETAIL
...