Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 658
Filtrar
1.
Front Immunol ; 15: 1404384, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38953035

RESUMEN

Introduction: Schistosomiasis (SM) is a parasitic disease caused by Schistosoma mansoni. SM causes chronic inflammation induced by parasitic eggs, with collagen/fibrosis deposition in the granuloma process in the liver, spleen, central nervous system, kidneys, and lungs. Pulmonary arterial hypertension (PAH) is a clinical manifestation characterized by high pressure in the pulmonary circulation and right ventricular overload. This study investigated the production of functional autoantibodies (fAABs) against the second loop of the G-protein-coupled receptor (GPCR) in the presence of hepatic and PAH forms of human SM. Methods: Uninfected and infected individuals presenting acute and chronic manifestations (e.g., hepatointestinal, hepato-splenic without PAH, and hepato-splenic with PAH) of SM were clinically evaluated and their blood was collected to identify fAABs/GPCRs capable of recognizing endothelin 1, angiotensin II, and a-1 adrenergic receptor. Human serum was analyzed in rat cardiomyocytes cultured in the presence of the receptor antagonists urapidil, losartan, and BQ123. Results: The fAABs/GPCRs from chronic hepatic and PAH SM individuals, but not from acute SM individuals, recognized the three receptors. In the presence of the antagonists, there was a reduction in beating rate changes in cultured cardiomyocytes. In addition, binding sites on the extracellular domain functionality of fAABs were identified, and IgG1 and/or IgG3 antibodies were found to be related to fAABs. Conclusion: Our data suggest that fAABs against GPCR play an essential role in vascular activity in chronic SM (hepatic and PAH) and might be involved in the development of hypertensive forms of SM.


Asunto(s)
Autoanticuerpos , Receptores Acoplados a Proteínas G , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Humanos , Animales , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Ratas , Masculino , Femenino , Adulto , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/etiología , Persona de Mediana Edad , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/parasitología , Esquistosomiasis mansoni/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis/inmunología
2.
Front Immunol ; 15: 1372957, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779688

RESUMEN

Background: Schistosomiasis is a common cause of pulmonary hypertension (PH) worldwide. Type 2 inflammation contributes to the development of Schistosoma-induced PH. Specifically, interstitial macrophages (IMs) derived from monocytes play a pivotal role by producing thrombospondin-1 (TSP-1), which in turn activates TGF-ß, thereby driving the pathology of PH. Resident and recruited IM subpopulations have recently been identified. We hypothesized that in Schistosoma-PH, one IM subpopulation expresses monocyte recruitment factors, whereas recruited monocytes become a separate IM subpopulation that expresses TSP-1. Methods: Mice were intraperitoneally sensitized and then intravenously challenged with S. mansoni eggs. Flow cytometry on lungs and blood was performed on wildtype and reporter mice to identify IM subpopulations and protein expression. Single-cell RNA sequencing (scRNAseq) was performed on flow-sorted IMs from unexposed and at day 1, 3 and 7 following Schistosoma exposure to complement flow cytometry based IM characterization and identify gene expression. Results: Flow cytometry and scRNAseq both identified 3 IM subpopulations, characterized by CCR2, MHCII, and FOLR2 expression. Following Schistosoma exposure, the CCR2+ IM subpopulation expanded, suggestive of circulating monocyte recruitment. Schistosoma exposure caused increased monocyte-recruitment ligand CCL2 expression in the resident FOLR2+ IM subpopulation. In contrast, the vascular pathology-driving protein TSP-1 was greatest in the CCR2+ IM subpopulation. Conclusion: Schistosoma-induced PH involves crosstalk between IM subpopulations, with increased expression of monocyte recruitment ligands by resident FOLR2+ IMs, and the recruitment of CCR2+ IMs which express TSP-1 that activates TGF-ß and causes PH.


Asunto(s)
Hipertensión Pulmonar , Macrófagos , Animales , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/parasitología , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/patología , Ratones , Macrófagos/inmunología , Macrófagos/parasitología , Fenotipo , Schistosoma mansoni/inmunología , Ratones Endogámicos C57BL , Esquistosomiasis/inmunología , Esquistosomiasis/complicaciones , Esquistosomiasis/parasitología , Modelos Animales de Enfermedad , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/complicaciones , Esquistosomiasis mansoni/patología , Trombospondina 1/genética , Trombospondina 1/metabolismo , Monocitos/inmunología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Femenino , Schistosoma/inmunología , Schistosoma/fisiología , Pulmón/inmunología , Pulmón/parasitología , Pulmón/patología
3.
Front Immunol ; 13: 929552, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36263057

RESUMEN

Schistosomiasis is a neglected tropical disease caused by worms of the genus Schistosoma spp. The progression of disease results in intense tissue fibrosis and high mortality rate. After egg deposition by adult worms, the inflammatory response is characterized by the robust activation of type 2 immunity. Monocytes and macrophages play critical roles during schistosomiasis. Inflammatory Ly6Chigh monocytes are recruited from the blood to the inflammatory foci and differentiate into alternatively activated macrophages (AAMs), which promote tissue repair. The common chain of ß2-integrins (CD18) regulates monocytopoiesis and mediates resistance to experimental schistosomiasis. There is still limited knowledge about mechanisms controlled by CD18 that impact monocyte development and effector cells such as macrophages during schistosomiasis. Here, we show that CD18low mice chronically infected with S. mansoni display monocyte progenitors with reduced proliferative capacity, resulting in the accumulation of the progenitor cell denominated proliferating-monocyte (pMo). Consequently, inflammatory Ly6Chigh and patrolling Ly6Clow monocytes are reduced in the bone marrow and blood. Mechanistically, low CD18 expression decreases Irf8 gene expression in pMo progenitor cells, whose encoded transcription factor regulates CSFR1 (CD115) expression on the cell surface. Furthermore, low CD18 expression affects the accumulation of inflammatory Ly6Chigh CD11b+ monocytes in the liver while the adoptive transference of these cells to infected-CD18low mice reduced the inflammatory infiltrate and fibrosis in the liver. Importantly, expression of Il4, Chil3l3 and Arg1 was downregulated, CD206+PD-L2+ AAMs were reduced and there were lower levels of IL-10 in the liver of CD18low mice chronically infected with S. mansoni. Overall, these findings suggest that CD18 controls the IRF8-CD115 axis on pMo progenitor cells, affecting their proliferation and maturation of monocytes. At the same time, CD18 is crucial for the appropriate polarization and function of AAMs and tissue repair during chronic schistosomiasis.


Asunto(s)
Antígenos CD18 , Esquistosomiasis , Animales , Ratones , Fibrosis , Integrinas/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Macrófagos , Monocitos , Esquistosomiasis/inmunología , Antígenos CD18/metabolismo
4.
PLoS Pathog ; 18(2): e1010233, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35108342

RESUMEN

Schistosomiasis, which is caused by infection with Schistosoma spp., is characterized by granuloma and fibrosis in response to egg deposition. Pattern recognition receptors are important to sense invading Schistosoma, triggering an innate immune response, and subsequently shaping adaptive immunity. Cyclic GMP-AMP synthase (cGAS) was identified as a major cytosolic DNA sensor, which catalyzes the formation of cyclic GMP-AMP (cGAMP), a critical second messenger for the activation of the adaptor protein stimulator of interferon genes (STING). The engagement of STING by cGAMP leads to the activation of TANK-binding kinase 1 (TBK1), interferon regulatory factor 3 (IRF3), and the subsequent type I interferon (IFN) response. cGAS is suggested to regulate infectious diseases, autoimmune diseases, and cancer. However, the function of cGAS in helminth infection is unclear. In this study, we found that Cgas deficiency enhanced the survival of mice infected with S. japonicum markedly, without affecting the egg load in the liver. Consistently, Cgas deletion alleviated liver pathological impairment, reduced egg granuloma formation, and decreased fibrosis severity. In contrast, Sting deletion reduced the formation of egg granulomas markedly, but not liver fibrosis. Notably, Cgas or Sting deficiency reduced the production of IFNß drastically in mice infected with S. japonicum. Intriguingly, intravenous administration of recombinant IFNß exacerbated liver damage and promoted egg granuloma formation, without affecting liver fibrosis. Clodronate liposome-mediated depletion of macrophages indicated that macrophages are the major type of cells contributing to the induction of the type I IFN response during schistosome infection. Moreover, cGAS is important for type I IFN production and phosphorylation of TBK1 and IRF3 in response to stimulation with S. japonicum egg- or adult worm-derived DNA in macrophages. Our results clarified the immunomodulatory effect of cGAS in the regulation of liver granuloma formation during S. japonicum infection, involving sensing schistosome-derived DNA and producing type I IFN. Additionally, we showed that cGAS regulates liver fibrosis in a STING-type I-IFN-independent manner.


Asunto(s)
Interferón Tipo I/inmunología , Proteínas de la Membrana/inmunología , Nucleotidiltransferasas/inmunología , Esquistosomiasis Japónica/inmunología , Esquistosomiasis/inmunología , Esquistosomiasis/parasitología , Animales , Femenino , Inmunidad , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nucleotidiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
5.
PLoS Pathog ; 17(12): e1010064, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34969052

RESUMEN

Schistosomes are long lived, intravascular parasitic platyhelminths that infect >200 million people globally. The molecular mechanisms used by these blood flukes to dampen host immune responses are described in this review. Adult worms express a collection of host-interactive tegumental ectoenzymes that can cleave host signaling molecules such as the "alarmin" ATP (cleaved by SmATPDase1), the platelet activator ADP (SmATPDase1, SmNPP5), and can convert AMP into the anti-inflammatory mediator adenosine (SmAP). SmAP can additionally cleave the lipid immunomodulator sphingosine-1-phosphate and the proinflammatory anionic polymer, polyP. In addition, the worms release a barrage of proteins (e.g., SmCB1, SjHSP70, cyclophilin A) that can impinge on immune cell function. Parasite eggs also release their own immunoregulatory proteins (e.g., IPSE/α1, omega1, SmCKBP) as do invasive cercariae (e.g., Sm16, Sj16). Some schistosome glycans (e.g., LNFPIII, LNnT) and lipids (e.g., Lyso-PS, LPC), produced by several life stages, likewise affect immune cell responses. The parasites not only produce eicosanoids (e.g., PGE2, PGD2-that can be anti-inflammatory) but can also induce host cells to release these metabolites. Finally, the worms release extracellular vesicles (EVs) containing microRNAs, and these too have been shown to skew host cell metabolism. Thus, schistosomes employ an array of biomolecules-protein, lipid, glycan, nucleic acid, and more, to bend host biochemistry to their liking. Many of the listed molecules have been individually shown capable of inducing aspects of the polarized Th2 response seen following infection (with the generation of regulatory T cells (Tregs), regulatory B cells (Bregs) and anti-inflammatory, alternatively activated (M2) macrophages). Precisely how host cells integrate the impact of these myriad parasite products following natural infection is not known. Several of the schistosome immunomodulators described here are in development as novel therapeutics against autoimmune, inflammatory, and other, nonparasitic, diseases.


Asunto(s)
Proteínas del Helminto/inmunología , Factores Inmunológicos/inmunología , Schistosoma/inmunología , Esquistosomiasis/inmunología , Animales , Humanos
6.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34281269

RESUMEN

The host-parasite schistosome relationship relies heavily on the interplay between the strategies imposed by the schistosome worm and the defense mechanisms the host uses to counter the line of attack of the parasite. The ultimate goal of the schistosome parasite entails five important steps: evade elimination tactics, survive within the human host, develop into adult forms, propagate in large numbers, and transmit from one host to the next. The aim of the parasitized host on the other hand is either to cure or limit infection. Therefore, it is a battle between two conflicting aspirations. From the host's standpoint, infection accompanies a plethora of immunological consequences; some are set in place to defend the host, while most end up promoting chronic disease, which ultimately crosses paths with oxidative stress and cancer. Understanding these networks provides attractive opportunities for anti-schistosome therapeutic development. Hence, this review discusses the mechanisms by which schistosomes modulate the human immune response with ultimate links to oxidative stress and genetic instability.


Asunto(s)
Citocinas/metabolismo , Interacciones Huésped-Parásitos/inmunología , Esquistosomiasis/inmunología , Esquistosomiasis/metabolismo , Animales , Linfocitos B Reguladores/inmunología , Basófilos/inmunología , Células Dendríticas/inmunología , Eosinófilos/inmunología , Humanos , Macrófagos/inmunología , Mastocitos/inmunología , MicroARNs/inmunología , Modelos Inmunológicos , Estrés Oxidativo , Schistosoma/inmunología , Schistosoma/patogenicidad , Esquistosomiasis/parasitología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología
7.
Front Immunol ; 12: 635513, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33953712

RESUMEN

Schistosome infection is a major cause of global morbidity, particularly in sub-Saharan Africa. However, there is no effective vaccine for this major neglected tropical disease, and re-infection routinely occurs after chemotherapeutic treatment. Following invasion through the skin, larval schistosomula enter the circulatory system and migrate through the lung before maturing to adulthood in the mesenteric or urogenital vasculature. Eggs released from adult worms can become trapped in various tissues, with resultant inflammatory responses leading to hepato-splenic, intestinal, or urogenital disease - processes that have been extensively studied in recent years. In contrast, although lung pathology can occur in both the acute and chronic phases of schistosomiasis, the mechanisms underlying pulmonary disease are particularly poorly understood. In chronic infection, egg-mediated fibrosis and vascular destruction can lead to the formation of portosystemic shunts through which eggs can embolise to the lungs, where they can trigger granulomatous disease. Acute schistosomiasis, or Katayama syndrome, which is primarily evident in non-endemic individuals, occurs during pulmonary larval migration, maturation, and initial egg-production, often involving fever and a cough with an accompanying immune cell infiltrate into the lung. Importantly, lung migrating larvae are not just a cause of inflammation and pathology but are a key target for future vaccine design. However, vaccine efforts are hindered by a limited understanding of what constitutes a protective immune response to larvae. In this review, we explore the current understanding of pulmonary immune responses and inflammatory pathology in schistosomiasis, highlighting important unanswered questions and areas for future research.


Asunto(s)
Enfermedades Pulmonares Parasitarias/parasitología , Pulmón/parasitología , Schistosoma/patogenicidad , Esquistosomiasis/parasitología , Animales , Modelos Animales de Enfermedad , Interacciones Huésped-Parásitos , Humanos , Evasión Inmune , Pulmón/efectos de los fármacos , Pulmón/inmunología , Enfermedades Pulmonares Parasitarias/inmunología , Enfermedades Pulmonares Parasitarias/prevención & control , Ratones , Vacunas Antiprotozoos/uso terapéutico , Schistosoma/efectos de los fármacos , Schistosoma/inmunología , Esquistosomiasis/inmunología , Esquistosomiasis/prevención & control , Esquistosomicidas/uso terapéutico
8.
Front Immunol ; 12: 635985, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746974

RESUMEN

Despite mass drug administration programmes with praziquantel, the prevalence of schistosomiasis remains high. A vaccine is urgently needed to control transmission of this debilitating disease. As some promising schistosomiasis vaccine candidates are moving through pre-clinical and clinical testing, we review the immunological challenges that these vaccine candidates may encounter in transitioning through the clinical trial phases in endemic settings. Prior exposure of the target population to schistosomes and other infections may impact vaccine response and efficacy and therefore requires considerable attention. Schistosomes are known for their potential to induce T-reg/IL-10 mediated immune suppression in populations which are chronically infected. Moreover, endemicity of schistosomiasis is focal whereby target and trial populations may exhibit several degrees of prior exposure as well as in utero exposure which may increase heterogeneity of vaccine responses. The age dependent distribution of exposure and development of acquired immunity, and general differences in the baseline immunological profile, adds to the complexity of selecting suitable trial populations. Similarly, prior or concurrent infections with other parasitic helminths, viral and bacterial infections, may alter immunological responses. Consequently, treatment of co-infections may benefit the immunogenicity of vaccines and may be considered despite logistical challenges. On the other hand, viral infections leave a life-long immunological imprint on the human host. Screening for serostatus may be needed to facilitate interpretation of vaccine responses. Co-delivery of schistosome vaccines with PZQ is attractive from a perspective of implementation but may complicate the immunogenicity of schistosomiasis vaccines. Several studies have reported PZQ treatment to induce both transient and long-term immuno-modulatory effects as a result of tegument destruction, worm killing and subsequent exposure of worm antigens to the host immune system. These in turn may augment or antagonize vaccine immunogenicity. Understanding the complex immunological interactions between vaccine, co-infections or prior exposure is essential in early stages of clinical development to facilitate phase 3 clinical trial design and implementation policies. Besides well-designed studies in different target populations using schistosome candidate vaccines or other vaccines as models, controlled human infections could also help identify markers of immune protection in populations with different disease and immunological backgrounds.


Asunto(s)
Desarrollo de Medicamentos , Enfermedades Endémicas/prevención & control , Vacunas Antiprotozoos/uso terapéutico , Schistosoma/inmunología , Esquistosomiasis/prevención & control , Animales , Coinfección , Diseño de Fármacos , Interacciones Huésped-Parásitos , Humanos , Inmunogenicidad Vacunal , Praziquantel/uso terapéutico , Vacunas Antiprotozoos/efectos adversos , Vacunas Antiprotozoos/inmunología , Schistosoma/patogenicidad , Esquistosomiasis/epidemiología , Esquistosomiasis/inmunología , Esquistosomiasis/transmisión , Esquistosomicidas/uso terapéutico
9.
Curr Top Med Chem ; 21(3): 193-204, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32972342

RESUMEN

Schistosomiasis is a neglected tropical disease. It is related to long-lasting granulomatous fibrosis and inflammation of target organs, and current sub-optimal pharmacological treatment creates global public health concerns. Intravascular worms and eggs release antigens and extracellular vesicles that target host endothelial cells, modulate the immune system, and stimulate the release of damageassociated molecular patterns (DAMPs). ATP, one of the most studied DAMPs, triggers a cascade of autocrine and paracrine actions through purinergic P2X and P2Y receptors, which are shaped by ectonucleotidases (CD39). Both P2 receptor families, and in particular P2Y1, P2Y2, P2Y12, and P2X7 receptors, have been attracting increasing interest in several inflammatory diseases and drug development. Current data obtained from the murine model unveiled a CD39-ADP-P2Y1/P2Y12 receptors signaling pathway linked to the liver and mesenteric exacerbations of schistosomal inflammation. Therefore, we proposed that members of this purinergic signaling could be putative pharmacological targets to reduce schistosomal morbidity.


Asunto(s)
Antihelmínticos/farmacología , Receptores Purinérgicos/inmunología , Esquistosomiasis/tratamiento farmacológico , Animales , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Schistosoma/efectos de los fármacos , Schistosoma/inmunología , Esquistosomiasis/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
10.
Immunology ; 162(2): 123-134, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32614982

RESUMEN

Macrophages are fundamental to sustain physiological equilibrium and to regulate the pathogenesis of parasitic and metabolic processes. The functional heterogeneity and immune responses of macrophages are shaped by cellular metabolism in response to the host's intrinsic factors, environmental cues and other stimuli during disease. Parasite infections induce a complex cascade of cytokines and metabolites that profoundly remodel the metabolic status of macrophages. In particular, helminths polarize macrophages to an M2 state and induce a metabolic shift towards reliance on oxidative phosphorylation, lipid oxidation and amino acid metabolism. Accumulating data indicate that helminth-induced activation and metabolic reprogramming of macrophages underlie improvement in overall whole-body metabolism, denoted by improved insulin sensitivity, body mass in response to high-fat diet and atherogenic index in mammals. This review aims to highlight the metabolic changes that occur in human and murine-derived macrophages in response to helminth infections and helminth products, with particular interest in schistosomiasis and soil-transmitted helminths.


Asunto(s)
Helmintiasis/inmunología , Helmintos/inmunología , Intestinos/inmunología , Intestinos/parasitología , Macrófagos/inmunología , Schistosoma/inmunología , Esquistosomiasis/inmunología , Animales , Citocinas/inmunología , Humanos , Macrófagos/parasitología
11.
Curr Drug Discov Technol ; 18(4): 473-484, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32767945

RESUMEN

Schistosome infection is regarded as one of the most important and neglected tropical diseases associated with poor sanitation. Like other living organisms, schistosomes employ multiple biological processes, of which some are regulated by a post-translational modification called Adenosine Diphosphate-ribosylation (ADP-ribosylation), catalyzed by ADP-ribosyltransferases. ADP-ribosylation is the addition of ADP-ribose moieties from Nicotinamide Adenine Dinucleotide (NAD+) to various targets, which include proteins and nucleotides. It is crucial in biological processes such as DNA repair, apoptosis, carbohydrate metabolism and catabolism. In the absence of a vaccine against schistosomiasis, this becomes a promising pathway in the identification of drug targets against various forms of this infection. The tegument of the worm is an encouraging immunogenic target for anti-schistosomal vaccine development. Vaccinology, molecular modeling and target-based drug discovery strategies have been used for years in drug discovery and for vaccine development. In this paper, we outline ADP-ribosylation and other different approaches to drug discovery and vaccine development against schistosomiasis.


Asunto(s)
ADP-Ribosilación/inmunología , Antihelmínticos/farmacología , Enfermedades Desatendidas/terapia , Schistosoma/inmunología , Esquistosomiasis/terapia , ADP-Ribosilación/efectos de los fármacos , Animales , Antihelmínticos/uso terapéutico , Antígenos Helmínticos/inmunología , Descubrimiento de Drogas/métodos , Humanos , Enfermedades Desatendidas/inmunología , Enfermedades Desatendidas/parasitología , Schistosoma/efectos de los fármacos , Esquistosomiasis/inmunología , Esquistosomiasis/parasitología , Desarrollo de Vacunas/métodos
12.
Front Immunol ; 11: 609994, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33281832

RESUMEN

Blood flukes of the genus Schistosoma are covered by a protective heptalaminated, double lipid bilayer surface membrane. Large amounts of sphingomyelin (SM) in the outer leaflet form with surrounding water molecules a tight hydrogen bond barrier, which allows entry of nutrients and prevents access of host immune effectors. Excessive hydrolysis of SM to phosphoryl choline and ceramide via activation of the parasite tegument-associated neutral sphingomyelinase (nSMase) with the polyunsaturated fatty acid, arachidonic acid (ARA) leads to parasite death, via allowing exposure of apical membrane antigens to antibody-dependent cell-mediated cytotoxicity (ADCC), and accumulation of the pro-apoptotic ceramide. Surface membrane nSMase represents, thus, a worm Achilles heel, and ARA a valid schistosomicide. Several experiments conducted in vitro using larval, juvenile, and adult Schistosoma mansoni and Schistosoma haematobium documented ARA schistosomicidal potential. Arachidonic acid schistosomicidal action was shown to be safe and efficacious in mice and hamsters infected with S. mansoni and S. haematobium, respectively, and in children with light S. mansoni infection. A combination of praziquantel and ARA led to outstanding cure rates in children with heavy S. mansoni infection. Additionally, ample evidence was obtained for the powerful ARA ovocidal potential in vivo and in vitro against S. mansoni and S. haematobium liver and intestine eggs. Studies documented ARA as an endogenous schistosomicide in the final mammalian and intermediate snail hosts, and in mice and hamsters, immunized with the cysteine peptidase-based vaccine. These findings together support our advocating the nutrient ARA as the safe and efficacious schistosomicide of the future.


Asunto(s)
Antígenos Helmínticos/administración & dosificación , Ácido Araquidónico/uso terapéutico , Proteasas de Cisteína/administración & dosificación , Schistosoma/efectos de los fármacos , Esquistosomiasis/tratamiento farmacológico , Esquistosomicidas/uso terapéutico , Vacunas/administración & dosificación , Animales , Antígenos Helmínticos/inmunología , Ácido Araquidónico/efectos adversos , Ácido Araquidónico/metabolismo , Proteasas de Cisteína/inmunología , Modelos Animales de Enfermedad , Interacciones Huésped-Parásitos , Humanos , Recuento de Huevos de Parásitos , Schistosoma/inmunología , Schistosoma/patogenicidad , Esquistosomiasis/inmunología , Esquistosomiasis/metabolismo , Esquistosomiasis/parasitología , Esquistosomicidas/efectos adversos , Resultado del Tratamiento , Vacunación , Vacunas/inmunología
13.
Parasit Vectors ; 13(1): 334, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32611373

RESUMEN

BACKGROUND: The main symptoms of schistosomiasis are granuloma and fibrosis, caused by Schistosoma eggs. Numerous types of cells and cytokines are involved in the progression of Schistosoma infection. As a class of innate immune cells, γδ T cells play critical roles in the early immune response. However, their role in modulating granuloma and fibrosis remains to be clarified. METHODS: Liver fibrosis in wild-type (WT) mice and T cell receptor (TCR) δ knockout (KO) mice infected with Schistosoma japonicum was examined via Masson's trichrome staining of collagen deposition and quantitative reverse transcriptase-PCR (RT-PCR) of fibrosis-related genes. Granuloma was detected by hematoxylin-eosin (H&E) staining and quantified. Flow cytometry was used for immune cell profiling and for detecting cytokine secretion. The abundance of the related cytokines was measured using quantitative RT-PCR. RESULTS: The livers of S. japonicum-infected mice had significantly increased proportions of interleukin (IL)-17A producing γδ T cells and secreted IL-17A. Compared with the WT mice, TCR δ deficiency resulted in reduced pathological impairment and fibrosis in the liver and increased survival in infected mice. In addition, the profibrogenic effects of γδ T cells in infected mice were associated with enhanced CD11b+Gr-1+ cells, concurrent with increased expression of transforming growth factor (TGF)-ß in the liver. CONCLUSIONS: In this mouse model of Schistosoma infection, γδ T cells may promote liver fibrosis by recruiting CD11b+Gr-1+ cells. These findings shed new light on the pathogenesis of liver pathology in murine schistosomiasis.


Asunto(s)
Interleucina-17/metabolismo , Hígado/patología , Esquistosomiasis Japónica , Linfocitos T/metabolismo , Animales , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Granuloma/parasitología , Granuloma/patología , Hígado/parasitología , Cirrosis Hepática/parasitología , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Schistosoma japonicum/inmunología , Schistosoma japonicum/patogenicidad , Esquistosomiasis/inmunología , Esquistosomiasis/patología , Esquistosomiasis Japónica/inmunología , Esquistosomiasis Japónica/patología , Subgrupos de Linfocitos T/metabolismo
14.
Front Immunol ; 11: 1018, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582161

RESUMEN

The deeply rooted, intricate relationship between the Schistosoma parasite and the human host has enabled the parasite to successfully survive within the host and surreptitiously evade the host's immune attacks. The parasite has developed a variety of strategies in its immunomodulatory armamentarium to promote infection without getting harmed or killed in the battlefield of immune responses. These include the production of immunomodulatory molecules, alteration of membranes, and the promotion of granuloma formation. Schistosomiasis thus serves as a paradigm for understanding the Th2 immune responses seen in various helminthiases. This review therefore aims to summarize the immunomodulatory mechanisms of the schistosome parasites to survive inside the host. Understanding these immunomodulatory strategies not only provides information on parasite-host interactions, but also forms the basis in the development of novel drugs and vaccines against the schistosome infection, as well as various types of autoimmune and inflammatory conditions.


Asunto(s)
Antihelmínticos/uso terapéutico , Antígenos Helmínticos/inmunología , Schistosoma/fisiología , Esquistosomiasis/inmunología , Células Th2/inmunología , Animales , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Inmunomodulación , Esquistosomiasis/terapia
15.
Infect Immun ; 88(8)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32341115

RESUMEN

The parasites and eggs of helminths, including schistosomes, are associated with factors that can modulate the nature and outcomes of host immune responses, particularly enhancing type 2 immunity and impairing the effects of type 1 and type 17 immunity. The main species of schistosomes that cause infection in humans are capable of generating a microenvironment that allows survival of the parasite by evasion of the immune response. Schistosome infections are associated with beneficial effects on chronic immune disorders, including allergies, autoimmune diseases, and alloimmune responses. Recently, there has been increasing research interest in the role of schistosomes in immunoregulation during human infection, and the mechanisms underlying these roles continue to be investigated. Further studies may identify potential opportunities to develop new treatments for immune disease. In this review, we provide an update on the advances in our understanding of schistosome-associated modulation of the cells of the innate and adaptive immune systems as well as the potential role of schistosome-associated factors as therapeutic modulators of immune disorders, including allergies, autoimmune diseases, and transplant immunopathology. We also discuss potential opportunities for targeting schistosome-induced immunoregulation for future translation to the clinical setting.


Asunto(s)
Enfermedades Autoinmunes/terapia , Hipersensibilidad/terapia , Factores Inmunológicos/uso terapéutico , Schistosoma japonicum/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis/terapia , Inmunidad Adaptativa/efectos de los fármacos , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/parasitología , Enfermedades Autoinmunes/patología , Hipersensibilidad/inmunología , Hipersensibilidad/parasitología , Hipersensibilidad/patología , Evasión Inmune , Inmunidad Innata/efectos de los fármacos , Inmunomodulación , Inmunoterapia/métodos , Trasplante de Órganos/rehabilitación , Schistosoma japonicum/química , Schistosoma mansoni/química , Esquistosomiasis/inmunología , Esquistosomiasis/parasitología , Esquistosomiasis/patología , Células TH1/inmunología , Células TH1/parasitología , Células Th17/inmunología , Células Th17/parasitología , Células Th2/inmunología , Células Th2/parasitología , Cigoto/química , Cigoto/inmunología
16.
Front Immunol ; 11: 61, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32132991

RESUMEN

The parasitic worms, Schistosoma mansoni and Schistosoma japonicum, reside in the mesenteric veins, where they release eggs that induce a dramatic granulomatous response in the liver and intestines. Subsequently, infection may further develop into significant fibrosis and portal hypertension. Over the past several years, uncovering the mechanism of immunopathology in schistosomiasis has become a major research objective. It is known that T lymphocytes, especially CD4+ T cells, are essential for immune responses against Schistosoma species. However, obtaining a clear understanding of how T lymphocytes regulate the pathological process is proving to be a daunting challenge. To date, CD4+ T cell subsets have been classified into several distinct T helper (Th) phenotypes including Th1, Th2, Th17, T follicular helper cells (Tfh), Th9, and regulatory T cells (Tregs). In the case of schistosomiasis, the granulomatous inflammation and the chronic liver pathology are critically regulated by the Th1/Th2 responses. Animal studies suggest that there is a moderate Th1 response to parasite antigens during the acute stage, but then, egg-derived antigens induce a sustained and dominant Th2 response that mediates granuloma formation and liver fibrosis. In addition, the newly discovered Th17 cells also play a critical role in the hepatic immunopathology of schistosomiasis. Within the liver, Tregs are recruited to hepatic granulomas and exert an immunosuppressive role to limit the granulomatous inflammation and fibrosis. Moreover, recent studies have shown that Tfh and Th9 cells might also promote liver granulomas and fibrogenesis in the murine schistosomiasis. Thus, during infection, T-cell subsets undergo complicated cross-talk with antigen presenting cells that then defines their various roles in the local microenvironment for regulating the pathological progression of schistosomiasis. This current review summarizes a vast body of literature to elucidate the contribution of T lymphocytes and their associated cytokines in the immunopathology of schistosomiasis.


Asunto(s)
Hígado/inmunología , Esquistosomiasis/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/inmunología , Granuloma/inmunología , Granuloma/parasitología , Humanos , Hígado/parasitología , Hígado/patología , Cirrosis Hepática/inmunología , Cirrosis Hepática/parasitología , Cirrosis Hepática/patología , Ratones , Schistosoma japonicum/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis/patología , Células T Auxiliares Foliculares/inmunología , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología
17.
Parasit Vectors ; 12(1): 383, 2019 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-31362766

RESUMEN

BACKGROUND: Schistosoma mekongi, which causes schistosomiasis in humans, is an important public health issue in Southeast Asia. Treatment with praziquantel is the primary method of control but emergence of praziquantel resistance requires the development of alternative drugs and vaccines. Calcium-dependent cysteine protease (calpain) is a novel vaccine candidate that has been studied in S. mansoni, S. japonicum, and protozoans including malaria, leishmania and trypanosomes. However, limited information is available on the properties and functions of calpain in other Schistosoma spp., including S. mekongi. In this study, we functionally characterized calpain 1 of S. mekongi (SmeCalp1). RESULTS: Calpain 1 of S. mekongi was obtained from transcriptomic analysis of S. mekongi; it had the highest expression level of all isoforms tested and was predominantly expressed in the adult male. SmeCalp1 cDNA is 2274 bp long and encodes 758 amino acids, with 85% to 90% homology with calpains in other Schistosoma species. Recombinant SmeCalp1 (rSmeCalp1), with a molecular weight of approximately 86.7 kDa, was expressed in bacteria and stimulated a marked antibody response in mice. Native SmeCalp1 was detected in crude worm extract and excretory-secretory product, and it was mainly localized in the tegument of the adult male; less signal was detected in the adult female worm. Thus, SmeCalp1 may play a role in surface membrane synthesis or host-parasite interaction. We assessed the protease activity of rSmeCalp1 and demonstrated that rSmeCalp1 could cleave the calpain substrate N-succinyl-Leu-Leu-Val-Tyr-7-amino-4-methylcoumarin, that was inhibited by calpain inhibitors (MDL28170 and E64c). Additionally, rSmeCalp1 could degrade the biological substrates fibronectin (blood clotting protein) and human complement C3, indicating important roles in the intravascular system and in host immune evasion. CONCLUSIONS: SmeCalp1 is expressed on the tegumental surface of the parasite and can cleave host defense molecules; thus, it might participate in growth, development and survival during the entire life-cycle of S. mekongi. Information on the properties and functions of SmeCalp1 reported herein will be advantageous in the development of effective drugs and vaccines against S. mekongi and other schistosomes.


Asunto(s)
Antígenos Helmínticos/inmunología , Calpaína/genética , Calpaína/metabolismo , Schistosoma/enzimología , Animales , Antígenos Helmínticos/genética , Cumarinas/metabolismo , Proteasas de Cisteína/genética , Proteasas de Cisteína/metabolismo , Femenino , Inmunización , Masculino , Ratones , Ratones Endogámicos ICR , Oligopéptidos/metabolismo , Schistosoma/genética , Esquistosomiasis/inmunología , Esquistosomiasis/parasitología , Análisis de Secuencia de ADN
18.
Front Immunol ; 10: 858, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191512

RESUMEN

Like soil-transmitted helminth infections, schistosomiasis is an important neglected tropical disease (NTD) related to poverty with a major impact on public health in developing countries. Diagnosis of active infection is crucial for surveillance of controlled or post-elimination schistosomiasis areas. In addition, the use of conventional diagnostic tools in non-exposed populations (such as travelers) results in misdiagnoses in the prepatent period of infection. Also, the accuracy of standard tests applied in low-endemicity areas (LEAs) decreases after several rounds of treatment. We aimed to determine whether it would be necessary to replace schistosomiasis conventional diagnostic tests such as parasitological methods in LEAs. Also, we evaluate the use of new tools in non-endemic areas. Reliable, cheap and easy-to-use diagnostic tools are needed to respond to the demands of a new era of elimination and eradication of schistosomiasis. To this end, molecular diagnosis-including nucleic acid-based assays (loop-mediated isothermal amplification, polymerase chain reaction) and circulating cathodic and anodic antigen detection tests have become promising strategies. In this review, we attempt to address the use of alternative diagnostic tests for active infection detection and drug-monitoring after specific schistosomiasis treatment.


Asunto(s)
Enfermedades Desatendidas/diagnóstico , Esquistosomiasis/diagnóstico , Antihelmínticos/uso terapéutico , Antígenos Helmínticos/análisis , Humanos , Sistemas de Atención de Punto , Praziquantel/uso terapéutico , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/inmunología
19.
Eur J Immunol ; 49(7): 1067-1081, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30919955

RESUMEN

Alternatively activated Mφs (AAMφ) accumulate in hepatic granulomas during schistosomiasis and have been suggested to originate in the bone marrow. What is less understood is how these Mφ responses are regulated after S. mansoni infection. Here, we investigated the role of IL-4 receptor α-chain (IL-4Rα)-signalling in the dynamics of liver Mφ responses. We observed that IL-4Rα signalling was dispensable for the recruitment of Ly6Chi monocytes and for their conversion into F4/80hi CD64hi CD11bhi Mφ. Moreover, while IL-4Rα provided an AAMφ phenotype to liver F4/80hi CD64hi CD11bhi Mφ that was associated with regulation of granuloma formation, it was dispensable for host survival. Resident F4/80hi CD64hi CD11blo Mφ did not upregulate the AAMφ signature gene Ym1. Rather, resident Mφ nearly disappeared by week 8 after infection and artificial ablation of resident Mφ in CD169DTR mice did not affect the response to S. mansoni infection. Interestingly, ablation of CD169+ cells in naive mice resulted in the accumulation of F4/80hi CD64hi CD11bhi Mφ, which was amplified when ablation occurred during schistosomiasis. Altogether, our results suggest the ablation of resident KCs after S. mansoni infection to be associated with the recruitment and accumulation of F4/80hi CD64hi CD11bhi Mφ with lyz2-dependent IL-4Rα contributing to the regulation of granuloma inflammation but being dispensable for host survival.


Asunto(s)
Granuloma/inmunología , Macrófagos del Hígado/inmunología , Hígado/patología , Macrófagos/inmunología , Receptores de Superficie Celular/metabolismo , Schistosoma mansoni/fisiología , Esquistosomiasis/inmunología , Técnicas de Ablación , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Superficie Celular/genética , Transducción de Señal
20.
Front Immunol ; 10: 26, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30761125

RESUMEN

Schistosomiasis is a major helminthic disease in which damage to the affected organs is orchestrated by a pathogenic host CD4 T helper (Th) cell-mediated immune response against parasite eggs. In the case of the species Schistosoma mansoni, the resulting granulomatous inflammation and fibrosis takes place in the liver and intestines. The magnitude of disease varies greatly from individual to individual but in a minority of patients, there is severe disease and death. S. mansoni infection in a murine model similarly results in marked strain variation of immunopathology. In the most commonly examined mouse strain, C57BL/6 (BL/6), there is relatively mild hepatic pathology arising in a Th2-dominated cytokine environment. In contrast, CBA mice develop decisively more severe lesions largely driven by proinflammatory IL-17-producing Th17 cells. Dendritic cells (DCs) from CBA mice differ sharply with those from BL/6 mice in that they vastly over-express the C-type lectin receptor (CLR) CD209a (SIGNR5), a homolog of human DC-SIGN, which senses glycans such as those produced by schistosome eggs. Silencing of CD209a, and recent studies with CD209a KO CBA mice have shown that this receptor is crucial to induce the pathogenic Th17 cell response; indeed, CD209a KO mice display markedly reduced immunopathology akin to that seen in BL/6 mice. Mechanistically, CD209a synergizes with the related CLRs Dectin-2 and Mincle to stimulate increased DC production of IL-1ß and IL-23, necessary for pathogenic Th17 cell development. These findings denote key molecular underpinnings of disease variability based on selection and function of contrasting Th cell subsets.


Asunto(s)
Interacciones Huésped-Parásitos/inmunología , Lectinas Tipo C/metabolismo , Esquistosomiasis/inmunología , Esquistosomiasis/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Biomarcadores , Moléculas de Adhesión Celular/metabolismo , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Unión Proteica , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptores de Superficie Celular/metabolismo , Schistosoma mansoni/inmunología , Esquistosomiasis/diagnóstico , Esquistosomiasis/parasitología , Índice de Severidad de la Enfermedad , Células Th2/inmunología , Células Th2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA