Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.025
Filtrar
1.
Expert Rev Proteomics ; 21(4): 205-216, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38584506

RESUMO

INTRODUCTION: Protein microarray is a promising immunomic approach for identifying biomarkers. Based on our previous study that reviewed parasite antigens and recent parasitic omics research, this article expands to include information on vector-borne parasitic diseases (VBPDs), namely, malaria, schistosomiasis, leishmaniasis, babesiosis, trypanosomiasis, lymphatic filariasis, and onchocerciasis. AREAS COVERED: We revisit and systematically summarize antigen markers of vector-borne parasites identified by the immunomic approach and discuss the latest advances in identifying antigens for the rational development of diagnostics and vaccines. The applications and challenges of this approach for VBPD control are also discussed. EXPERT OPINION: The immunomic approach has enabled the identification and/or validation of antigen markers for vaccine development, diagnosis, disease surveillance, and treatment. However, this approach presents several challenges, including limited sample size, variability in antigen expression, false-positive results, complexity of omics data, validation and reproducibility, and heterogeneity of diseases. In addition, antigen involvement in host immune evasion and antigen sensitivity/specificity are major issues in its application. Despite these limitations, this approach remains promising for controlling VBPD. Advances in technology and data analysis methods should continue to improve candidate antigen identification, as well as the use of a multiantigen approach in diagnostic and vaccine development for VBPD control.


Assuntos
Biomarcadores , Doenças Parasitárias , Humanos , Animais , Biomarcadores/sangue , Doenças Parasitárias/imunologia , Doenças Parasitárias/diagnóstico , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/imunologia , Análise Serial de Proteínas/métodos , Proteômica/métodos
2.
Biosci Rep ; 44(5)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38623843

RESUMO

Parasitic diseases are a serious global health concern, causing many common and severe infections, including Chagas disease, leishmaniasis, and schistosomiasis. The NLRP3 inflammasome belongs to the NLR (nucleotide-binding domain leucine-rich-repeat-containing proteins) family, which are cytosolic proteins playing key roles in the detection of pathogens. NLRP3 inflammasomes are activated in immune responses to Plasmodium, Leishmania, Toxoplasma gondii, Entamoeba histolytica, Trypanosoma cruzi, and other parasites. The role of NLRP3 is not fully understood, but it is a crucial component of the innate immune response to parasitic infections and its functions as a sensor triggering the inflammatory response to the invasive parasites. However, while this response can limit the parasites' growth, it can also result in potentially catastrophic host pathology. This makes it essential to understand how NLRP3 interacts with parasites to initiate the inflammatory response. Plasmodium hemozoin, Leishmania glycoconjugate lipophosphoglycan (LPG) and E. histolytica Gal/GalNAc lectin can stimulate NLRP3 activation, while the dense granule protein 9 (GRA9) of T. gondii has been shown to suppress it. Several other parasitic products also have diverse effects on NLRP3 activation. Understanding the mechanism of NLRP3 interaction with these products will help to develop advanced therapeutic approaches to treat parasitic diseases. This review summarizes current knowledge of the NLRP3 inflammasome's action on the immune response to parasitic infections and aims to determine the mechanisms through which parasitic molecules either activate or inhibit its action.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Inflamassomos/metabolismo , Inflamassomos/imunologia , Animais , Doenças Parasitárias/imunologia , Doenças Parasitárias/parasitologia , Doenças Parasitárias/metabolismo , Imunidade Inata
3.
Acta Trop ; 238: 106792, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36509129

RESUMO

Neglected tropical parasitic diseases (NTD) are prevalent in many countries and cost-effective treatments remain urgently needed. Novel approaches have been proposed to address these diseases through an action on immune co-inhibitory checkpoints which are exploited by parasites to evade the immune system. Among these checkpoints, TIM-3 has been shown to play a key role in antiparasitic immunity via a repression and functional attenuation of CD4+ and/or CD8+ T-cells. The present review discusses the role of the TIM-3/galectin-9 checkpoint in seven major NTD: Chagas disease, leishmaniasis and malaria (3 trypanosomatid infections), schistosomiasis, toxoplasmosis, echinococcosis and filariasis (4 helminth infections). In each case, the role of the checkpoint has been analyzed and the use of anti-TIM-3 antibodies evaluated as a potential therapeutic approach. In general, the parasitic infection is coupled with an upregulation of TIM-3 expressed on T cells, but not necessarily with an exhaustion of those T cells. In several cases, the use of anti-TIM-3 antibodies represent a possible strategy to reinforce the clearance and to reduce the parasite load. Promising data have been reported in cases of leishmaniasis, malaria and schistosomiasis, whereas a similar approach proved much less efficient (if not deleterious) in cases of echinococcosis and the Chagas disease. Nevertheless, the TIM-3 checkpoint warrants further consideration as a potential immune target to combat these pathologies, using antibodies or drugs capable of reducing directly or indirectly the expression and function of the checkpoint, to restore an immune control.


Assuntos
Galectinas , Receptor Celular 2 do Vírus da Hepatite A , Doenças Parasitárias , Animais , Humanos , Linfócitos T CD8-Positivos/imunologia , Galectinas/imunologia , Doenças Parasitárias/imunologia , Receptor Celular 2 do Vírus da Hepatite A/imunologia
4.
Sci Rep ; 12(1): 3197, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210503

RESUMO

The adaptive immune system is critical to an effective response to infection in vertebrates, with T-helper (Th) cells pivotal in orchestrating these responses. In natural populations where co-infections are the norm, different Th responses are likely to play an important role in maintaining host health and fitness, a relationship which remains poorly understood in wild animals. In this study, we characterised variation in functionally distinct Th responses in a wild population of Soay sheep by enumerating cells expressing Th-subset specific transcription factors and quantifying Th-associated cytokines. We tested the prediction that raised Th1 and Th2 responses should predict reduced apicomplexan and helminth parasite burdens, respectively. All measures of Th-associated cytokine production increased with age, while Th17- and regulatory Th-associated cytokine production increased more rapidly with age in males than females. Independent of age, sex, and each other, IL-4 and Gata3 negatively predicted gastro-intestinal nematode faecal egg count, while IFN-γ negatively predicted coccidian faecal oocyst count. Our results provide important support from outside the laboratory that Th1 and Th2 responses predict resistance to different kinds of parasites, and illustrate how harnessing specific reagents and tools from laboratory immunology will illuminate our understanding of host-parasite interactions in the wild.


Assuntos
Parasitos/imunologia , Doenças Parasitárias/imunologia , Ovinos/sangue , Ovinos/imunologia , Ovinos/parasitologia , Linfócitos T Auxiliares-Indutores/imunologia , Imunidade Adaptativa , Animais , Citocinas/sangue , Fezes/parasitologia , Feminino , Fator de Transcrição GATA3/sangue , Fator de Transcrição GATA3/metabolismo , Interações Hospedeiro-Parasita , Interleucina-4/sangue , Masculino , Doenças Parasitárias/parasitologia , Fenótipo , Prognóstico , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia , Fatores de Transcrição/sangue
5.
Biomolecules ; 12(2)2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-35204759

RESUMO

Human Leukocyte Antigen-G (HLA-G), a polymorphic non-classical HLA (HLA-Ib) with immune-regulatory properties in cancers and infectious diseases, presents both membrane-bound and soluble (sHLA-G) isoforms. Polymorphism has implications in host responses to pathogen infections and in pathogenesis. Differential expression patterns of HLA-G/sHLA-G or its polymorphism seem to be related to different pathological conditions, potentially acting as a disease progression biomarker. Pathogen antigens might be involved in the regulation of both membrane-bound and sHLA-G levels and impact immune responses during co-infections. The upregulation of HLA-G in viral and bacterial infections induce tolerance to infection. Recently, sHLA-G was found useful to identify the prognosis of Coronavirus disease 2019 (COVID-19) among patients and it was observed that the high levels of sHLA-G are associated with worse prognosis. The use of pathogens, such as Plasmodium falciparum, as immune modulators for other infections could be extended for the modulation of membrane-bound HLA-G in COVID-19-infected tissues. Overall, such information might open new avenues concerning the effect of some pathogens such as parasites in decreasing the expression level of HLA-G to restrict pathogenesis in some infections or to influence the immune responses after vaccination among others.


Assuntos
COVID-19/imunologia , Antígenos HLA-G/imunologia , Antígenos HLA-G/metabolismo , Imunomodulação , Doenças Parasitárias/imunologia , COVID-19/terapia , Humanos , Imunoterapia , Doenças Parasitárias/terapia
6.
J Immunol ; 208(2): 221-226, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35017211

RESUMO

Maternal infection during pregnancy is known to alter the development and function of offspring's immune system, leading to inappropriate immune responses to common childhood infections and immunizations. Although this is an expanding field, maternal parasitic infections remain understudied. Millions of women of reproductive age are currently at risk for parasitic infection, whereas many pregnant, chronically infected women are excluded from mass drug administration due partially to a lack of resources, as well as fear of unknown adverse fetal developmental outcomes. In areas endemic for multiple parasitic infections, such as sub-Saharan Africa, there are increased rates of morbidity and mortality for various infections during early childhood in comparison with nonendemic areas. Despite evidence supporting similar immunomodulatory effects between various parasite species, there is no clear mechanistic understanding of how maternal infection reprograms offspring immunity. This brief review will compare the effects of selected maternal parasitic infections on offspring immunity.


Assuntos
Desenvolvimento Fetal/imunologia , Helmintíase/imunologia , Malária Falciparum/imunologia , Doenças Parasitárias/transmissão , Complicações Parasitárias na Gravidez/epidemiologia , Adulto , África Subsaariana/epidemiologia , Animais , Feminino , Helmintíase/parasitologia , Helmintíase/transmissão , Helmintos/patogenicidade , Humanos , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Doenças Parasitárias/epidemiologia , Doenças Parasitárias/imunologia , Gravidez , Solo/parasitologia
7.
Int J Mol Sci ; 22(24)2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34948112

RESUMO

The human host immune responses to parasitic infections are complex. They can be categorized into four immunological pathways mounted against four types of parasitic infections. For intracellular protozoa, the eradicable host immunological pathway is TH1 immunity involving macrophages (M1), interferon gamma (IFNγ) CD4 T cells, innate lymphoid cells 1 (NKp44+ ILC1), CD8 T cells (Effector-Memory4, EM4), invariant natural killer T cells 1 (iNKT1) cells, and immunoglobulin G3 (IgG3) B cells. For intracellular protozoa, the tolerable host immunological pathway is TH1-like immunity involving macrophages (M2), interferon gamma (IFNγ)/TGFß CD4 T cells, innate lymphoid cells 1 (NKp44- ILC1), CD8 T cells (EM3), invariant natural killer T 1 (iNKT1) cells, and immunoglobulin A1 (IgA1) B cells. For free-living extracellular protozoa, the eradicable host immunological pathway is TH22 immunity involving neutrophils (N1), interleukin-22 CD4 T cells, innate lymphoid cells 3 (NCR+ ILC3), iNKT17 cells, and IgG2 B cells. For free-living extracellular protozoa, the tolerable host immunological pathway is TH17 immunity involving neutrophils (N2), interleukin-17 CD4 T cells, innate lymphoid cells 3 (NCR- ILC3), iNKT17 cells, and IgA2 B cells. For endoparasites (helminths), the eradicable host immunological pathway is TH2a immunity with inflammatory eosinophils (iEOS), interleukin-5/interleukin-4 CD4 T cells, interleukin-25 induced inflammatory innate lymphoid cells 2 (iILC2), tryptase-positive mast cells (MCt), iNKT2 cells, and IgG4 B cells. For ectoparasites (parasitic insects and arachnids), the eradicable host immunological pathway is TH2b immunity with inflammatory basophils, chymase- and tryptase-positive mast cells (MCct), interleukin-3/interleukin-4 CD4 T cells, interleukin-33 induced nature innate lymphoid cells 2 (nILC2), iNKT2 cells, and immunoglobulin E (IgE) B cells. The tolerable host immunity against ectoparasites and endoparasites is TH9 immunity with regulatory eosinophils, regulatory basophils, interleukin-9 mast cells (MMC9), thymic stromal lymphopoietin induced innate lymphoid cells 2, interleukin-9 CD4 T cells, iNKT2 cells, and IgA2 B cells. In addition, specific transcription factors important for specific immune responses were listed. This JAK/STAT signaling is key to controlling or inducing different immunological pathways. In sum, Tfh is related to STAT5ß, and BCL6 expression. Treg is related to STAT5α, STAT5ß, and FOXP3. TH1 immunity is related to STAT1α, STAT4, and T-bet. TH2a immunity is related to STAT6, STAT1α, GATA1, and GATA3. TH2b immunity is related to STAT6, STAT3, GATA2, and GATA3. TH22 immunity is associated with both STAT3α and AHR. THαß immunity is related to STAT1α, STAT1ß, STAT2, STAT3ß, and ISGF. TH1-like immunity is related to STAT1α, STAT4, STAT5α, and STAT5ß. TH9 immunity is related to STAT6, STAT5α, STAT5ß, and PU.1. TH17 immunity is related to STAT3α, STAT5α, STAT5ß, and RORG. TH3 immunity is related to STAT1α, STAT1ß, STAT2, STAT3ß, STAT5α, STAT5ß, and ISGF. This categorization provides a complete framework of immunological pathways against four types of parasitic infections. This framework as well as relevant JAK/STAT signaling can provide useful knowledge to control allergic hypersensitivities and parasitic infections via development of vaccines or drugs in the near future.


Assuntos
Citocinas/imunologia , Imunidade Inata , Leucócitos/imunologia , Doenças Parasitárias/imunologia , Humanos
8.
PLoS Comput Biol ; 17(12): e1009714, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34932551

RESUMO

Hosts diverge widely in how, and how well, they defend themselves against infection and immunopathology. Why are hosts so heterogeneous? Both epidemiology and life history are commonly hypothesized to influence host immune strategy, but the relationship between immune strategy and each factor has commonly been investigated in isolation. Here, we show that interactions between life history and epidemiology are crucial for determining optimal immune specificity and sensitivity. We propose a demographically-structured population dynamics model, in which we explore sensitivity and specificity of immune responses when epidemiological risks vary with age. We find that variation in life history traits associated with both reproduction and longevity alters optimal immune strategies-but the magnitude and sometimes even direction of these effects depends on how epidemiological risks vary across life. An especially compelling example that explains previously-puzzling empirical observations is that depending on whether infection risk declines or rises at reproductive maturity, later reproductive maturity can select for either greater or lower immune specificity, potentially illustrating why studies of lifespan and immune variation across taxa have been inconclusive. Thus, the sign of selection on the life history-immune specificity relationship can be reversed in different epidemiological contexts. Drawing on published life history data from a variety of chordate taxa, we generate testable predictions for this facet of the optimal immune strategy. Our results shed light on the causes of the heterogeneity found in immune defenses both within and among species and the ultimate variability of the relationship between life history and immune specificity.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Modelos Biológicos , Parasitos , Doenças Parasitárias , Animais , Evolução Biológica , Humanos , Longevidade/imunologia , Parasitos/imunologia , Parasitos/patogenicidade , Doenças Parasitárias/epidemiologia , Doenças Parasitárias/imunologia , Doenças Parasitárias/parasitologia , Dinâmica Populacional , Reprodução
9.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34768828

RESUMO

Cell death is an essential immunological apparatus of host defense, but dysregulation of mutually inclusive cell deaths poses severe threats during microbial and parasitic infections leading to deleterious consequences in the pathological progression of infectious diseases. Nucleotide-binding oligomerization domain (NOD)-Leucine-rich repeats (LRR)-containing receptors (NLRs), also called nucleotide-binding oligomerization (NOD)-like receptors (NLRs), are major cytosolic pattern recognition receptors (PRRs), their involvement in the orchestration of innate immunity and host defense against bacteria, viruses, fungi and parasites, often results in the cleavage of gasdermin and the release of IL-1ß and IL-18, should be tightly regulated. NLRs are functionally diverse and tissue-specific PRRs expressed by both immune and non-immune cells. Beyond the inflammasome activation, NLRs are also involved in NF-κB and MAPK activation signaling, the regulation of type I IFN (IFN-I) production and the inflammatory cell death during microbial infections. Recent advancements of NLRs biology revealed its possible interplay with pyroptotic cell death and inflammatory mediators, such as caspase 1, caspase 11, IFN-I and GSDMD. This review provides the most updated information that caspase 8 skews the NLRP3 inflammasome activation in PANoptosis during pathogen infection. We also update multidimensional roles of NLRP12 in regulating innate immunity in a content-dependent manner: novel interference of NLRP12 on TLRs and NOD derived-signaling cascade, and the recently unveiled regulatory property of NLRP12 in production of type I IFN. Future prospects of exploring NLRs in controlling cell death during parasitic and microbial infection were highlighted.


Assuntos
Infecções/imunologia , Proteínas NLR/fisiologia , Doenças Parasitárias/imunologia , Animais , Morte Celular/imunologia , Interações entre Hospedeiro e Microrganismos , Interações Hospedeiro-Parasita , Humanos , Mediadores da Inflamação/metabolismo , Proteínas NLR/genética , Proteínas NLR/imunologia , Proteínas NLR/metabolismo , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais , Vírus/imunologia
11.
Front Immunol ; 12: 696003, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34177963

RESUMO

Antiviral, antibacterial, and antiparasitic drugs and vaccines are essential to maintaining the health of humans and animals. Yet, their production can be slow and expensive, and efficacy lost once pathogens mount resistance. Chicken immunoglobulin Y (IgY) is a highly conserved homolog of human immunoglobulin G (IgG) that has shown benefits and a favorable safety profile, primarily in animal models of human infectious diseases. IgY is fast-acting, easy to produce, and low cost. IgY antibodies can readily be generated in large quantities with minimal environmental harm or infrastructure investment by using egg-laying hens. We summarize a variety of IgY uses, focusing on their potential for the detection, prevention, and treatment of human and animal infections.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Infecções Bacterianas/tratamento farmacológico , Galinhas/imunologia , Imunoensaio , Imunoglobulinas/uso terapêutico , Doenças Parasitárias/tratamento farmacológico , Viroses/tratamento farmacológico , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Especificidade de Anticorpos , Infecções Bacterianas/diagnóstico , Infecções Bacterianas/imunologia , Infecções Bacterianas/virologia , Humanos , Imunoglobulinas/biossíntese , Imunoglobulinas/imunologia , Doenças Parasitárias/diagnóstico , Doenças Parasitárias/imunologia , Doenças Parasitárias/virologia , Valor Preditivo dos Testes , Viroses/diagnóstico , Viroses/imunologia , Viroses/virologia
12.
Nat Commun ; 12(1): 3371, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099671

RESUMO

The role of p53 in tumor suppression has been extensively studied and well-established. However, the role of p53 in parasitic infections and the intestinal type 2 immunity is unclear. Here, we report that p53 is crucial for intestinal type 2 immunity in response to the infection of parasites, such as Tritrichomonas muris and Nippostrongylus brasiliensis. Mechanistically, p53 plays a critical role in the activation of the tuft cell-IL-25-type 2 innate lymphoid cell circuit, partly via transcriptional regulation of Lrmp in tuft cells. Lrmp modulates Ca2+ influx and IL-25 release, which are critical triggers of type 2 innate lymphoid cell response. Our results thus reveal a previously unrecognized function of p53 in regulating intestinal type 2 immunity to protect against parasitic infections, highlighting the role of p53 as a guardian of immune integrity.


Assuntos
Imunidade Inata/imunologia , Intestinos/imunologia , Nippostrongylus/imunologia , Doenças Parasitárias/imunologia , Tritrichomonas/imunologia , Proteína Supressora de Tumor p53/imunologia , Animais , Linhagem Celular Tumoral , Eosinófilos/imunologia , Eosinófilos/parasitologia , Regulação da Expressão Gênica , Células Caliciformes/imunologia , Células Caliciformes/parasitologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/parasitologia , Intestinos/parasitologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nippostrongylus/fisiologia , Doenças Parasitárias/metabolismo , Doenças Parasitárias/parasitologia , Tritrichomonas/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
13.
Front Immunol ; 12: 661241, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122419

RESUMO

As a relatively successful pathogen, several parasites can establish long-term infection in host. This "harmonious symbiosis" status relies on the "precise" manipulation of host immunity and metabolism, however, the underlying mechanism is still largely elusive. Immunometabolism is an emerging crossed subject in recent years. It mainly discusses the regulatory mechanism of metabolic changes on reprogramming the key transcriptional and post-transcriptional events related to immune cell activation and effect, which provides a novel insight for understanding how parasites regulate the infection and immunity in hosts. The present study reviewed the current research progress on metabolic reprogramming mechanism exploited by parasites to modulate the function in various immune cells, highlighting the future exploitation of key metabolites or metabolic events to clarify the underlying mechanism of anti-parasite immunity and design novel intervention strategies against parasitic infection.


Assuntos
Células Dendríticas/imunologia , Linfócitos/imunologia , Macrófagos/imunologia , Doenças Parasitárias/imunologia , Plasmodium/imunologia , Schistosoma/imunologia , Trypanosoma/imunologia , Animais , Células Dendríticas/metabolismo , Células Dendríticas/parasitologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Linfócitos/metabolismo , Linfócitos/parasitologia , Macrófagos/metabolismo , Macrófagos/parasitologia , Doenças Parasitárias/metabolismo , Doenças Parasitárias/parasitologia , Plasmodium/fisiologia , Schistosoma/fisiologia , Trypanosoma/fisiologia
14.
Adv Protein Chem Struct Biol ; 125: 193-213, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33931139

RESUMO

Parasitic organisms of various genera have threatened humankind. Although they are not always fatal but can damage the well-being of an individual in terms of both economic and societal crisis. Marked progress has been made toward eliminating those pathogenic organisms, however, complete removal is still not possible. Several antiparasitic drug moieties have been largely commercialized and are routinely used at the same time novel drug candidates are still required. Programmed cell death (PCD) is a vital biological phenomenon inside every organism. Particularly, induction of the death signaling inside the parasitic species through selective targeting of effective drug candidates is one of the major strategies to combat these infectious organisms. In this chapter significance of apoptosis induction to eliminate the parasitic disease has been illustrated with suitable references. Moreover, we have shared our own experiences of apoptosis induction in eliminating a World Health Organization enlisted Neglected Tropical Disease, lymphatic filariasis. On the other hand, we have also tried to put some light on the mechanism of apoptosis in different parasites.


Assuntos
Antiparasitários/uso terapêutico , Apoptose , Desenvolvimento de Medicamentos , Doenças Parasitárias , Transdução de Sinais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Humanos , Doenças Parasitárias/tratamento farmacológico , Doenças Parasitárias/imunologia , Doenças Parasitárias/parasitologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
15.
Front Immunol ; 12: 675751, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34017345

RESUMO

Knowledge of glycogen synthase kinase 3ß (GSK3ß) activity and the molecules identified that regulate its function in infections caused by pathogenic microorganisms is crucial to understanding how the intensity of the inflammatory response can be controlled in the course of infections. In recent years many reports have described small molecular weight synthetic and natural compounds, proteins, and interference RNA with the potential to regulate the GSK3ß activity and reduce the deleterious effects of the inflammatory response. Our goal in this review is to summarize the most recent advances on the role of GSK3ß in the inflammatory response caused by bacteria, bacterial virulence factors (i.e. LPS and others), viruses, and parasites and how the regulation of its activity, mainly its inhibition by different type of molecules, modulates the inflammation.


Assuntos
Infecções Bacterianas/imunologia , Glicogênio Sintase Quinase 3 beta/fisiologia , Inflamação/etiologia , Doenças Parasitárias/imunologia , Viroses/imunologia , Animais , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Humanos , Fosforilação
16.
Plant Physiol ; 185(4): 1282-1291, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33793887

RESUMO

Parasitic plants pose a major biotic threat to plant growth and development and lead to losses in crop productivity of billions of USD annually. By comparison with "normal" autotrophic plants, parasitic plants live a heterotrophic lifestyle and rely on water, solutes and to a greater (holoparasitic plants) or lesser extent (hemiparasitic plants) on sugars from other host plants. Most hosts are unable to detect an infestation by plant parasites or unable to fend off these parasitic invaders. However, a few hosts have evolved defense strategies to avoid infestation or protect themselves actively post-attack often leading to full or partial resistance. Here, we review the current state of our understanding of the defense strategies to plant parasitism used by host plants with emphasis on the active molecular resistance mechanisms. Furthermore, we outline the perspectives and the potential of future studies that will be indispensable to develop and breed resistant crops.


Assuntos
Produtos Agrícolas/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Doenças Parasitárias/imunologia , Imunidade Vegetal/fisiologia , Virulência/fisiologia , Estados Unidos
17.
Infect Genet Evol ; 89: 104753, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33545392

RESUMO

B lymphocytes are primarily well known for their contribution to immunity by antibody production, antigen presentation and, the production of cytokines. In recent years several studies demonstrated the existence of B cells with regulatory functions, which have been termed regulatory B cells (Bregs), similar to regulatory T cells (Tregs). Bregs are a subpopulation of B cells that have immunosuppressive effects via the production of regulatory cytokines including interleukin-10 (IL-10), transforming growth factor-ß (TGF-ß), and IL-35. Bregs limit host defense against various pathogens. In addition, Bregs contribute to increased levels of regulatory cytokines and leads to an induction of suppressive Tregs, which exert broader suppressive functions against various pathogens. The high percentage of Bregs is positively associated with viral and bacterial load and can contribute to poor vaccine responses. Bregs can also facilitate pathogen survival at an early stage of infection, and subsequently cause increased severity of disease by inhibiting pro-inflammatory cytokine production, macrophage activation, and inflammatory T cells activation such as Th1, Th17, and Th22. Also, Bregs afford protection against the hyper-inflammatory response in parasitic infections. Here we review the central role of Bregs in many major bacterial and viral human infections, and provide an overview of the immunoregulatory mechanisms used by Bregs.


Assuntos
Linfócitos B Reguladores/imunologia , Infecções Bacterianas/imunologia , Doenças Parasitárias/imunologia , Viroses/imunologia , Humanos , Ativação Linfocitária
18.
Biomed Pharmacother ; 133: 111033, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33378946

RESUMO

For decades, glucocorticoids (GC) have been used to treat several inflammatory conditions, including chronic and autoimmune diseases, due to their potent anti-inflammatory properties. In the context of infectious diseases, the use of GCs may be effective as adjuvant to antibiotic therapy by controlling excessive inflammatory responses resulting in better outcome in some cases. However, the use of GCs has been associated with a vast number of side effects, including increased probability of immunosuppression and consequent risk of opportunistic infection. Glucocorticoid-induced leucine zipper (GILZ) and Annexin A1 (AnxA1) are GC-induced proteins intrinsically involved with the anti-inflammatory functions of GCs without the associated adverse metabolic effects. Recent studies have shown that these GC-proteins exhibit pro-resolving effects. An essential characteristic of pro-resolving molecules is their ability to coordinate the resolution of inflammation and promote host defense in most experimental models of infection. Although the role of GILZ and AnxA1 in the context of infectious diseases remain to be better explored, herein we provide an overview of the emerging functions of these GC-proteins obtained from pre-clinical models of infectious diseases.


Assuntos
Anexina A1/metabolismo , Anti-Inflamatórios/uso terapêutico , Infecções Bacterianas/tratamento farmacológico , Glucocorticoides/uso terapêutico , Inflamação/tratamento farmacológico , Doenças Parasitárias/tratamento farmacológico , Fatores de Transcrição/metabolismo , Viroses/tratamento farmacológico , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Doenças Parasitárias/imunologia , Doenças Parasitárias/metabolismo , Doenças Parasitárias/parasitologia , Indução de Remissão , Transdução de Sinais , Resultado do Tratamento , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
20.
Front Immunol ; 11: 1805, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193291

RESUMO

Parasites, bacteria, and viruses pose serious threats to public health. Many parasite infections, including infections of protozoa and helminths, can inhibit inflammatory responses and impact disease outcomes caused by viral, bacterial, or other parasitic infections. Type I interferon (IFN-I) has been recognized as an essential immune effector in the host defense against various pathogens. In addition, IFN-I responses induced by co-infections with different pathogens may vary according to the host genetic background, immune status, and pathogen burden. However, there is only limited information on the roles of IFN-I in co-infections with parasites and viruses, bacteria, or other parasites. This review summarizes some recent findings on the roles of IFN-I in co-infections with parasites, including Leishmania spp., Plasmodium spp., Eimeria maxima, Heligmosomoides polygyrus, Brugia malayi, or Schistosoma mansoni, and viruses or bacteria and co-infections with different parasites (such as co-infection with Neospora caninum and Toxoplasma gondii, and co-infection with Plasmodium spp. and H. polygyrus). The potential mechanisms of host responses associated with co-infections, which may provide targets for immune intervention and therapies of the co-infections, are also discussed.


Assuntos
Bactérias/imunologia , Infecções Bacterianas/imunologia , Coinfecção , Interferon Tipo I/imunologia , Parasitos/imunologia , Doenças Parasitárias/imunologia , Viroses/imunologia , Vírus/imunologia , Animais , Bactérias/patogenicidade , Infecções Bacterianas/metabolismo , Infecções Bacterianas/terapia , Infecções Bacterianas/virologia , Interações Hospedeiro-Parasita , Humanos , Interferon Tipo I/metabolismo , Parasitos/patogenicidade , Doenças Parasitárias/metabolismo , Doenças Parasitárias/parasitologia , Doenças Parasitárias/terapia , Transdução de Sinais , Viroses/metabolismo , Viroses/terapia , Viroses/virologia , Vírus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...