Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Annu Rev Immunol ; 39: 639-665, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33646858

RESUMO

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


Assuntos
Helmintíase , Helmintos , Parasitos , Animais , Interações Hospedeiro-Parasita , Humanos , Imunidade Inata , Linfócitos , Linfócitos T
2.
Proc Natl Acad Sci U S A ; 119(36): e2202795119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36037362

RESUMO

Parasitic helminth infections, while a major cause of neglected tropical disease burden, negatively correlate with the incidence of immune-mediated inflammatory diseases such as inflammatory bowel diseases (IBD). To evade expulsion, helminths have developed sophisticated mechanisms to regulate their host's immune responses. Controlled experimental human helminth infections have been assessed clinically for treating inflammatory conditions; however, such a radical therapeutic modality has challenges. An alternative approach is to harness the immunomodulatory properties within the worm's excretory-secretory (ES) complement, its secretome. Here, we report a biologics discovery and validation pipeline to generate and screen in vivo a recombinant cell-free secretome library of helminth-derived immunomodulatory proteins. We successfully expressed 78 recombinant ES proteins from gastrointestinal hookworms and screened the crude in vitro translation reactions for anti-IBD properties in a mouse model of acute colitis. After statistical filtering and ranking, 20 proteins conferred significant protection against various parameters of colitis. Lead candidates from distinct protein families, including annexins, transthyretins, nematode-specific retinol-binding proteins, and SCP/TAPS were identified. Representative proteins were produced in mammalian cells and further validated, including ex vivo suppression of inflammatory cytokine secretion by T cells from IBD patient colon biopsies. Proteins identified herein offer promise as novel, safe, and mechanistically differentiated biologics for treating the globally increasing burden of inflammatory diseases.


Assuntos
Anti-Inflamatórios , Produtos Biológicos , Colite , Proteínas de Helminto , Doenças Inflamatórias Intestinais , Animais , Anti-Inflamatórios/farmacologia , Produtos Biológicos/farmacologia , Colite/tratamento farmacológico , Proteínas de Helminto/genética , Proteínas de Helminto/farmacologia , Helmintos , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/parasitologia , Camundongos
3.
Immunity ; 43(5): 998-1010, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26522986

RESUMO

Intestinal helminths are potent regulators of their host's immune system and can ameliorate inflammatory diseases such as allergic asthma. In the present study we have assessed whether this anti-inflammatory activity was purely intrinsic to helminths, or whether it also involved crosstalk with the local microbiota. We report that chronic infection with the murine helminth Heligmosomoides polygyrus bakeri (Hpb) altered the intestinal habitat, allowing increased short chain fatty acid (SCFA) production. Transfer of the Hpb-modified microbiota alone was sufficient to mediate protection against allergic asthma. The helminth-induced anti-inflammatory cytokine secretion and regulatory T cell suppressor activity that mediated the protection required the G protein-coupled receptor (GPR)-41. A similar alteration in the metabolic potential of intestinal bacterial communities was observed with diverse parasitic and host species, suggesting that this represents an evolutionary conserved mechanism of host-microbe-helminth interactions.


Assuntos
Microbioma Gastrointestinal/imunologia , Helmintos/imunologia , Hipersensibilidade/imunologia , Inflamação/imunologia , Inflamação/parasitologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Adulto , Idoso , Animais , Asma/imunologia , Asma/microbiologia , Asma/parasitologia , Citocinas/imunologia , Ácidos Graxos/imunologia , Feminino , Humanos , Hipersensibilidade/microbiologia , Hipersensibilidade/parasitologia , Inflamação/microbiologia , Mucosa Intestinal/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Nematospiroides dubius/imunologia , Receptores Acoplados a Proteínas G/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/microbiologia , Infecções por Strongylida/parasitologia
4.
PLoS Pathog ; 16(5): e1008508, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32407385

RESUMO

Parasitic helminths have coevolved with humans over millennia, intricately refining and developing an array of mechanisms to suppress or skew the host's immune system, thereby promoting their long-term survival. Some helminths, such as hookworms, cause little to no overt pathology when present in modest numbers and may even confer benefits to their human host. To exploit this evolutionary phenomenon, clinical trials of human helminth infection have been established and assessed for safety and efficacy for a range of immune dysfunction diseases and have yielded mixed outcomes. Studies of live helminth therapy in mice and larger animals have convincingly shown that helminths and their excretory/secretory products possess anti-inflammatory drug-like properties and represent an untapped pharmacopeia. These anti-inflammatory moieties include extracellular vesicles, proteins, glycans, post-translational modifications, and various metabolites. Although the concept of helminth-inspired therapies holds promise, it also presents a challenge to the drug development community, which is generally unfamiliar with foreign biologics that do not behave like antibodies. Identification and characterization of helminth molecules and vesicles and the molecular pathways they target in the host present a unique opportunity to develop tailored drugs inspired by nature that are efficacious, safe, and have minimal immunogenicity. Even so, much work remains to mine and assess this out-of-the-box therapeutic modality. Industry-based organizations need to consider long-haul investments aimed at unraveling and exploiting unique and differentiated mechanisms of action as opposed to toe-dipping entries with an eye on rapid and profitable turnarounds.


Assuntos
Proteínas de Helminto/imunologia , Helmintíase/imunologia , Helmintos/imunologia , Imunomodulação , Animais , Helmintíase/patologia , Helmintíase/terapia , Helmintos/patogenicidade , Humanos
5.
Nat Immunol ; 11(12): 1093-101, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20953201

RESUMO

Regulatory T cells (T(reg) cells) have a critical role in the maintenance of immunological self-tolerance. Here we show that treatment of naive human or mouse T cells with IL-35 induced a regulatory population, which we call 'iT(R)35 cells', that mediated suppression via IL-35 but not via the inhibitory cytokines IL-10 or transforming growth factor-ß (TGF-ß). We found that iT(R)35 cells did not express or require the transcription factor Foxp3, and were strongly suppressive and stable in vivo. T(reg) cells induced the generation of iT(R)35 cells in an IL-35- and IL-10-dependent manner in vitro and induced their generation in vivo under inflammatory conditions in intestines infected with Trichuris muris and within the tumor microenvironment (B16 melanoma and MC38 colorectal adenocarcinoma), where they contributed to the regulatory milieu. Thus, iT(R)35 cells constitute a key mediator of infectious tolerance and contribute to T(reg) cell-mediated tumor progression. Furthermore, iT(R)35 cells generated ex vivo might have therapeutic utility.


Assuntos
Tolerância Imunológica/imunologia , Interleucinas/imunologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Interleucinas/metabolismo , Camundongos , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/citologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo
6.
Nat Immunol ; 10(7): 697-705, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19465906

RESUMO

Dendritic cells can prime naive CD4+ T cells; however, here we demonstrate that dendritic cell-mediated priming was insufficient for the development of T helper type 2 cell-dependent immunity. We identify basophils as a dominant cell population that coexpressed major histocompatibility complex class II and interleukin 4 message after helminth infection. Basophilia was promoted by thymic stromal lymphopoietin, and depletion of basophils impaired immunity to helminth infection. Basophils promoted antigen-specific CD4+ T cell proliferation and interleukin 4 production in vitro, and transfer of basophils augmented the population expansion of helminth-responsive CD4+ T cells in vivo. Collectively, our studies suggest that major histocompatibility complex class II-dependent interactions between basophils and CD4+ T cells promote T helper type 2 cytokine responses and immunity to helminth infection.


Assuntos
Basófilos/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade/imunologia , Animais , Basófilos/citologia , Basófilos/metabolismo , Antígeno CD11c/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/imunologia , Proliferação de Células , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Immunoblotting , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia , Células Th2/metabolismo , Timo/citologia , Timo/imunologia , Tricuríase/imunologia , Tricuríase/parasitologia
7.
J Immunol ; 203(10): 2724-2734, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31586037

RESUMO

Alternatively activated macrophages are essential effector cells during type 2 immunity and tissue repair following helminth infections. We previously showed that Ym1, an alternative activation marker, can drive innate IL-1R-dependent neutrophil recruitment during infection with the lung-migrating nematode, Nippostrongylus brasiliensis, suggesting a potential role for the inflammasome in the IL-1-mediated innate response to infection. Although inflammasome proteins such as NLRP3 have important proinflammatory functions in macrophages, their role during type 2 responses and repair are less defined. We therefore infected Nlrp3 -/- mice with N. brasiliensis Unexpectedly, compared with wild-type (WT) mice, infected Nlrp3 -/- mice had increased neutrophilia and eosinophilia, correlating with enhanced worm killing but at the expense of increased tissue damage and delayed lung repair. Transcriptional profiling showed that infected Nlrp3 -/- mice exhibited elevated type 2 gene expression compared with WT mice. Notably, inflammasome activation was not evident early postinfection with N. brasiliensis, and in contrast to Nlrp3 -/- mice, antihelminth responses were unaffected in caspase-1/11-deficient or WT mice treated with the NLRP3-specific inhibitor MCC950. Together these data suggest that NLRP3 has a role in constraining lung neutrophilia, helminth killing, and type 2 immune responses in an inflammasome-independent manner.


Assuntos
Inflamassomos/fisiologia , Pneumopatias Parasitárias/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Nippostrongylus/imunologia , Infecções por Strongylida/imunologia , Animais , Caspase 1/fisiologia , Quimiotaxia de Leucócito , Eosinofilia/etiologia , Eosinofilia/imunologia , Furanos/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis , Imunidade Inata , Indenos , Interleucina-4/farmacologia , Lectinas/biossíntese , Lectinas/genética , Pulmão/patologia , Pulmão/fisiologia , Pneumopatias Parasitárias/complicações , Pneumopatias Parasitárias/patologia , Pneumopatias Parasitárias/fisiopatologia , Macrófagos Alveolares/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neutrófilos/imunologia , Regeneração , Infecções por Strongylida/complicações , Infecções por Strongylida/patologia , Infecções por Strongylida/fisiopatologia , Sulfonamidas/farmacologia , Sulfonas , Transcrição Gênica , beta-N-Acetil-Hexosaminidases/biossíntese , beta-N-Acetil-Hexosaminidases/genética
8.
Nature ; 504(7478): 153-7, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24185009

RESUMO

The development and severity of inflammatory bowel diseases and other chronic inflammatory conditions can be influenced by host genetic and environmental factors, including signals derived from commensal bacteria. However, the mechanisms that integrate these diverse cues remain undefined. Here we demonstrate that mice with an intestinal epithelial cell (IEC)-specific deletion of the epigenome-modifying enzyme histone deacetylase 3 (HDAC3(ΔIEC) mice) exhibited extensive dysregulation of IEC-intrinsic gene expression, including decreased basal expression of genes associated with antimicrobial defence. Critically, conventionally housed HDAC3(ΔIEC) mice demonstrated loss of Paneth cells, impaired IEC function and alterations in the composition of intestinal commensal bacteria. In addition, HDAC3(ΔIEC) mice showed significantly increased susceptibility to intestinal damage and inflammation, indicating that epithelial expression of HDAC3 has a central role in maintaining intestinal homeostasis. Re-derivation of HDAC3(ΔIEC) mice into germ-free conditions revealed that dysregulated IEC gene expression, Paneth cell homeostasis and intestinal barrier function were largely restored in the absence of commensal bacteria. Although the specific mechanisms through which IEC-intrinsic HDAC3 expression regulates these complex phenotypes remain to be determined, these data indicate that HDAC3 is a critical factor that integrates commensal-bacteria-derived signals to calibrate epithelial cell responses required to establish normal host-commensal relationships and maintain intestinal homeostasis.


Assuntos
Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Homeostase , Mucosa Intestinal/enzimologia , Intestinos/microbiologia , Simbiose , Adulto , Animais , Bactérias/genética , Colite Ulcerativa/enzimologia , Colite Ulcerativa/genética , Colite Ulcerativa/microbiologia , Doença de Crohn/enzimologia , Doença de Crohn/genética , Doença de Crohn/microbiologia , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Histona Desacetilases/genética , Humanos , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , RNA Ribossômico 16S/genética , Transdução de Sinais
9.
Genes Dev ; 25(23): 2480-8, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22156208

RESUMO

Macrophages, a key cellular component of inflammation, become functionally polarized in a signal- and context-specific manner. Th2 cytokines such as interleukin 4 (IL-4) polarize macrophages to a state of alternative activation that limits inflammation and promotes wound healing. Alternative activation is mediated by a transcriptional program that is influenced by epigenomic modifications, including histone acetylation. Here we report that macrophages lacking histone deacetylase 3 (HDAC3) display a polarization phenotype similar to IL-4-induced alternative activation and, furthermore, are hyperresponsive to IL-4 stimulation. Throughout the macrophage genome, HDAC3 deacetylates histone tails at regulatory regions, leading to repression of many IL-4-regulated genes characteristic of alternative activation. Following exposure to Schistosoma mansoni eggs, a model of Th2 cytokine-mediated disease that is limited by alternative activation, pulmonary inflammation was ameliorated in mice lacking HDAC3 in macrophages. Thus, HDAC3 functions in alternative activation as a brake whose release could be of benefit in the treatment of multiple inflammatory diseases.


Assuntos
Epigênese Genética , Histona Desacetilases/genética , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Animais , Histona Desacetilases/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos , Pneumonia/enzimologia , Pneumonia/imunologia , Pneumonia/parasitologia , Schistosoma mansoni , Células Th2/imunologia , Células Th2/metabolismo
10.
J Infect Dis ; 218(9): 1511-1516, 2018 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-29462492

RESUMO

Helminth infections in children are associated with impaired cognitive development; however, the biological mechanisms for this remain unclear. Using a murine model of gastrointestinal helminth infection, we demonstrate that early-life exposure to helminths promotes local and systemic inflammatory responses and transient changes in the gastrointestinal microbiome. Behavioral and cognitive analyses performed 9-months postinfection revealed deficits in spatial recognition memory and an anxiety-like behavioral phenotype in worm-infected mice, which was associated with neuropathology and increased microglial activation within the brain. This study demonstrates a previously unrecognized mechanism through which helminth infections may influence cognitive function, via perturbations in the gut-immune-brain axis.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/parasitologia , Trato Gastrointestinal/parasitologia , Helmintíase/complicações , Animais , Ansiedade/parasitologia , Modelos Animais de Doenças , Helmintíase/parasitologia , Helmintos/patogenicidade , Masculino , Transtornos da Memória/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Neuropatologia/métodos
11.
PLoS Pathog ; 11(10): e1005209, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26485648

RESUMO

Infection with the human liver fluke Opisthorchis viverrini induces cancer of the bile ducts, cholangiocarcinoma (CCA). Injury from feeding activities of this parasite within the human biliary tree causes extensive lesions, wounds that undergo protracted cycles of healing, and re-injury over years of chronic infection. We show that O. viverrini secreted proteins accelerated wound resolution in human cholangiocytes, an outcome that was compromised following silencing of expression of the fluke-derived gene encoding the granulin-like growth factor, Ov-GRN-1. Recombinant Ov-GRN-1 induced angiogenesis and accelerated mouse wound healing. Ov-GRN-1 was internalized by human cholangiocytes and induced gene and protein expression changes associated with wound healing and cancer pathways. Given the notable but seemingly paradoxical properties of liver fluke granulin in promoting not only wound healing but also a carcinogenic microenvironment, Ov-GRN-1 likely holds marked potential as a therapeutic wound-healing agent and as a vaccine against an infection-induced cancer of major public health significance in the developing world.


Assuntos
Carcinogênese/metabolismo , Proteínas de Helminto/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Opistorquíase/complicações , Opisthorchis/metabolismo , Cicatrização/fisiologia , Sequência de Aminoácidos , Animais , Neoplasias dos Ductos Biliares/parasitologia , Colangiocarcinoma/parasitologia , Humanos , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Opistorquíase/metabolismo , Progranulinas , Interferência de RNA
12.
J Infect Dis ; 211(3): 416-25, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25139017

RESUMO

Na-ASP-2 is an efficacious hookworm vaccine antigen. However, despite elucidation of its crystal structure and studies addressing its immunobiology, the function of Na-ASP-2 has remained elusive. We probed a 9000-protein human proteome microarray with Na-ASP-2 and showed binding to CD79A, a component of the B-cell antigen receptor complex. Na-ASP-2 bound to human B lymphocytes ex vivo and downregulated the transcription of approximately 1000 B-cell messenger RNAs (mRNAs), while only approximately 100 mRNAs were upregulated, compared with control-treated cells. The expression of a range of molecules was affected by Na-ASP-2, including factors involved in leukocyte transendothelial migration pathways and the B-cell signaling receptor pathway. Of note was the downregulated transcription of lyn and pi3k, molecules that are known to interact with CD79A and control B-cell receptor signaling processes. Together, these results highlight a previously unknown interaction between a hookworm-secreted protein and B cells, which has implications for helminth-driven immunomodulation and vaccine development. Further, the novel use of human protein microarrays to identify host-pathogen interactions, coupled with ex vivo binding studies and subsequent analyses of global gene expression in human host cells, demonstrates a new pipeline by which to explore the molecular basis of infectious diseases.


Assuntos
Ancylostomatoidea/imunologia , Linfócitos B/imunologia , Infecções por Uncinaria/imunologia , Receptores de Células Precursoras de Linfócitos B/imunologia , Proteoma/imunologia , Proteínas Recombinantes/imunologia , Transdução de Sinais/imunologia , Adulto , Animais , Antígenos de Helmintos/imunologia , Antígenos CD79/imunologia , Células Cultivadas , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Proteínas de Helminto/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Leucócitos Mononucleares/imunologia , Pessoa de Meia-Idade , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Análise Serial de Proteínas/métodos , Proteoma/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Transdução de Sinais/genética , Transcrição Gênica/genética , Transcrição Gênica/imunologia , Regulação para Cima/genética , Regulação para Cima/imunologia , Quinases da Família src/genética , Quinases da Família src/imunologia
13.
J Immunol ; 190(5): 2292-300, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23355735

RESUMO

Resistin-like molecule (RELM)α belongs to a family of secreted mammalian proteins that have putative immunomodulatory functions. Recent studies have identified a pathogenic role for RELMα in chemically induced colitis through effects on innate cell populations. However, whether RELMα regulates intestinal adaptive immunity to enteric pathogens is unknown. In this study, we employed Citrobacter rodentium as a physiologic model of pathogenic Escherichia coli-induced diarrheal disease, colitis, and Th17 cell responses. In response to Citrobacter, RELMα expression was induced in intestinal epithelial cells, infiltrating macrophages, and eosinophils of the infected colons. Citrobacter-infected RELMα(-/-) mice exhibited reduced infection-induced intestinal inflammation, characterized by decreased leukocyte recruitment to the colons and reduced immune cell activation compared with wild-type (WT) mice. Interestingly, Citrobacter colonization and clearance were unaffected in RELMα(-/-) mice, suggesting that the immune stimulatory effects of RELMα following Citrobacter infection were pathologic rather than host-protective. Furthermore, infected RELMα(-/-) mice exhibited decreased CD4(+) T cell expression of the proinflammatory cytokine IL-17A. To directly test whether RELMα promoted Citrobacter-induced intestinal inflammation via IL-17A, infected WT and IL-17A(-/-) mice were treated with rRELMα. RELMα treatment of Citrobacter-infected WT mice exacerbated intestinal inflammation and IL-17A expression whereas IL-17A(-/-) mice were protected from RELMα-induced intestinal inflammation. Finally, infected RELMα(-/-) mice exhibited reduced levels of serum IL-23p19 compared with WT mice, and RELMα(-/-) peritoneal macrophages showed deficient IL-23p19 induction. Taken together, these data identify a proinflammatory role for RELMα in bacterial-induced colitis and suggest that the IL-23/Th17 axis is a critical mediator of RELMα-induced inflammation.


Assuntos
Citrobacter rodentium/imunologia , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interleucina-17/imunologia , Intestinos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Imunidade Adaptativa/efeitos dos fármacos , Animais , Citrobacter rodentium/patogenicidade , Sulfato de Dextrana , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/microbiologia , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Interleucina-17/deficiência , Interleucina-17/genética , Subunidade p19 da Interleucina-23/sangue , Subunidade p19 da Interleucina-23/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Células Th17/imunologia , Células Th17/patologia
14.
J Immunol ; 189(9): 4371-8, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23024277

RESUMO

CD4(+) Th2 cytokine responses promote the development of allergic inflammation and are critical for immunity to parasitic helminth infection. Recent studies highlighted that basophils can promote Th2 cytokine-mediated inflammation and that phenotypic and functional heterogeneity exists between classical IL-3-elicited basophils and thymic stromal lymphopoietin (TSLP)-elicited basophils. However, whether distinct basophil populations develop after helminth infection and their relative contributions to anti-helminth immune responses remain to be defined. After Trichinella spiralis infection of mice, we show that basophil responses are rapidly induced in multiple tissue compartments, including intestinal-draining lymph nodes. Trichinella-induced basophil responses were IL-3-IL-3R independent but critically dependent on TSLP-TSLPR interactions. Selective depletion of basophils after Trichinella infection impaired infection-induced CD4(+) Th2 cytokine responses, suggesting that TSLP-dependent basophils augment Th2 cytokine responses after helminth infection. The identification and functional classification of TSLP-dependent basophils in a helminth infection model, coupled with their recently described role in promoting atopic dermatitis, suggests that these cells may be a critical population in promoting Th2 cytokine-associated inflammation in a variety of inflammatory or infectious settings. Collectively, these data suggest that the TSLP-basophil pathway may represent a new target in the design of therapeutic intervention strategies to promote or limit Th2 cytokine-dependent immunity and inflammation.


Assuntos
Basófilos/imunologia , Citocinas/biossíntese , Mucosa Intestinal/imunologia , Células Th2/imunologia , Timo/imunologia , Trichinella spiralis/imunologia , Triquinelose/imunologia , Animais , Basófilos/parasitologia , Basófilos/patologia , Citocinas/fisiologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/parasitologia , Mucosa Intestinal/parasitologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Estromais/imunologia , Células Estromais/parasitologia , Células Estromais/patologia , Células Th2/parasitologia , Células Th2/patologia , Timo/parasitologia , Timo/patologia , Triquinelose/metabolismo , Triquinelose/patologia , Linfopoietina do Estroma do Timo
15.
PLoS Negl Trop Dis ; 17(11): e0011695, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37956181

RESUMO

BACKGROUND: Trichuris trichiura (whipworm) is one of the most prevalent soil transmitted helminths (STH) affecting 604-795 million people worldwide. Diagnostic tools that are affordable and rapid are required for detecting STH. Here, we assessed the performance of the near-infrared spectroscopy (NIRS) technique coupled with machine learning algorithms to detect Trichuris muris in faecal, blood, serum samples and non-invasively through the skin of mice. METHODOLOGY: We orally infected 10 mice with 30 T. muris eggs (low dose group), 10 mice with 200 eggs (high dose group) and 10 mice were used as the control group. Using the NIRS technique, we scanned faecal, serum, whole blood samples and mice non-invasively through their skin over a period of 6 weeks post infection. Using artificial neural networks (ANN) and spectra of faecal, serum, blood and non-invasive scans from one experiment, we developed 4 algorithms to differentiate infected from uninfected mice. These models were validated on mice from a second independent experiment. PRINCIPAL FINDINGS: NIRS and ANN differentiated mice into the three groups as early as 2 weeks post infection regardless of the sample used. These results correlated with those from concomitant serological and parasitological investigations. SIGNIFICANCE: To our knowledge, this is the first study to demonstrate the potential of NIRS as a diagnostic tool for human STH infections. The technique could be further developed for large scale surveillance of soil transmitted helminths in human populations.


Assuntos
Helmintíase , Helmintos , Tricuríase , Humanos , Animais , Camundongos , Trichuris , Espectroscopia de Luz Próxima ao Infravermelho , Tricuríase/epidemiologia , Helmintíase/epidemiologia , Solo/parasitologia , Algoritmos , Fezes/parasitologia
16.
Nat Commun ; 14(1): 4503, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37495576

RESUMO

The reduced prevalence of insulin resistance and type 2 diabetes in countries with endemic parasitic worm infections suggests a protective role for worms against metabolic disorders, however clinical evidence has been non-existent. This 2-year randomised, double-blinded clinical trial in Australia of hookworm infection in 40 male and female adults at risk of type 2 diabetes assessed the safety and potential metabolic benefits of treatment with either 20 (n = 14) or 40 (n = 13) Necator americanus larvae (L3) or Placebo (n = 13) (Registration ACTRN12617000818336). Primary outcome was safety defined by adverse events and completion rate. Homoeostatic model assessment of insulin resistance, fasting blood glucose and body mass were key secondary outcomes. Adverse events were more frequent in hookworm-treated participants, where 44% experienced expected gastrointestinal symptoms, but completion rates were comparable to Placebo. Fasting glucose and insulin resistance were lowered in both hookworm-treated groups at 1 year, and body mass was reduced after L3-20 treatment at 2 years. This study suggests hookworm infection is safe in people at risk of type 2 diabetes and associated with improved insulin resistance, warranting further exploration of the benefits of hookworms on metabolic health.


Assuntos
Diabetes Mellitus Tipo 2 , Infecções por Uncinaria , Resistência à Insulina , Animais , Masculino , Feminino , Infecções por Uncinaria/complicações , Infecções por Uncinaria/tratamento farmacológico , Infecções por Uncinaria/epidemiologia , Necator americanus , Jejum
17.
Front Med (Lausanne) ; 9: 934852, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36186812

RESUMO

A decline in the prevalence of parasites such as hookworms appears to be correlated with the rise in non-communicable inflammatory conditions in people from high- and middle-income countries. This correlation has led to studies that have identified proteins produced by hookworms that can suppress inflammatory bowel disease (IBD) and asthma in animal models. Hookworms secrete a family of abundant netrin-domain containing proteins referred to as AIPs (Anti-Inflammatory Proteins), but there is no information on the structure-function relationships. Here we have applied a downsizing approach to the hookworm AIPs to derive peptides of 20 residues or less, some of which display anti-inflammatory effects when co-cultured with human peripheral blood mononuclear cells and oral therapeutic activity in a chemically induced mouse model of acute colitis. Our results indicate that a conserved helical region is responsible, at least in part, for the anti-inflammatory effects. This helical region has potential in the design of improved leads for treating IBD and possibly other inflammatory conditions.

18.
Transl Res ; 232: 88-102, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33676036

RESUMO

The symbiotic relationships shared between humans and their gastrointestinal parasites present opportunities to discover novel therapies for inflammatory diseases. A prime example of this phenomenon is the interaction of humans and roundworms such as the hookworm, Necator americanus. Epidemiological observations, animal studies and clinical trials using experimental human hookworm infection show that hookworms can suppress inflammation in a safe and well-tolerated way, and that the key to their immunomodulatory properties lies within their secreted proteome. Herein we describe the identification of 2 netrin domain-containing proteins from the N. americanus secretome, and explore their potential in treating intestinal inflammation in mouse models of ulcerative colitis. One of these proteins, subsequently named Na-AIP-1, was effective at suppressing disease when administered prophylactically in the acute TNBS-induced model of colitis. This protective effect was validated in the more robust CD4 T cell transfer model of chronic colitis, where prophylactic Na-AIP-1 reduced T-cell-dependent type-1 cytokine responses in the intestine and the associated intestinal pathology. Mechanistic studies revealed that depletion of CD11c+ cells abrogated the protective anticolitic effect of Na-AIP-1. Next generation sequencing of colon tissue in the T-cell transfer model of colitis revealed that Na-AIP-1 induced a transcriptomic profile associated with the downregulation of metabolic and signaling pathways involved in type-1 inflammation, notably TNF. Finally, co-culture of Na-AIP-1 with a human monocyte-derived M1 macrophage cell line resulted in significantly reduced secretion of TNF. Na-AIP-1 is now a candidate for clinical development as a novel therapeutic for the treatment of human inflammatory bowel diseases.


Assuntos
Anti-Inflamatórios/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Colite Ulcerativa/prevenção & controle , Proteínas de Helminto/administração & dosagem , Necator americanus/química , Netrinas/administração & dosagem , Animais , Linfócitos T CD4-Positivos/transplante , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/imunologia , Modelos Animais de Doenças , Feminino , Proteínas de Helminto/química , Proteínas de Helminto/genética , Infecções por Uncinaria/metabolismo , Humanos , Masculino , Inibidores de Metaloproteinases de Matriz/química , Camundongos Endogâmicos C57BL , Camundongos Knockout , Netrinas/análise , Proteínas Recombinantes/administração & dosagem
19.
Sci Rep ; 10(1): 13115, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32753607

RESUMO

Toxoplasmic encephalitis is an AIDS-defining condition. The decline of IFN-γ-producing CD4+ T cells in AIDS is a major contributing factor in reactivation of quiescent Toxoplasma gondii to an actively replicating stage of infection. Hence, it is important to characterize CD4-independent mechanisms that constrain acute T. gondii infection. We investigated the in vivo regulation of IFN-γ production by CD8+ T cells, DN T cells and NK cells in response to acute T. gondii infection. Our data show that processing of IFN-γ by these non-CD4 cells is dependent on both IL-12 and IL-18 and the secretion of bioactive IL-18 in response to T. gondii requires the sensing of viable parasites by multiple redundant inflammasome sensors in multiple hematopoietic cell types. Importantly, our results show that expansion of CD8+ T cells, DN T cells and NK cell by S4B6 IL-2 complex pre-treatment increases survival rates of mice infected with T. gondii and this is dependent on IL-12, IL-18 and IFN-γ. Increased survival is accompanied by reduced pathology but is independent of expansion of TReg cells or parasite burden. This provides evidence for a protective role of IL2C-mediated expansion of non-CD4 cells and may represent a promising lead to adjunct therapy for acute toxoplasmosis.


Assuntos
Interferon gama/biossíntese , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Interleucina-2/farmacologia , Toxoplasmose/imunologia , Toxoplasmose/prevenção & controle , Animais , Encéfalo/parasitologia , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Interleucina-2/química , Camundongos , Toxoplasma/efeitos dos fármacos , Toxoplasma/fisiologia , Toxoplasmose/metabolismo
20.
Clin Transl Gastroenterol ; 11(12): e00274, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33512796

RESUMO

INTRODUCTION: Celiac disease is an autoimmune disorder where intestinal immunopathology arises after gluten consumption. Previous studies suggested that hookworm infection restores gluten tolerance; however, these studies were small (n = 12) and not placebo controlled. METHODS: We undertook a randomized, placebo-controlled trial of hookworm infection in 54 people with celiac disease. The 94-week study involved treatment with either 20 or 40 Necator americanus third-stage larvae (L3-20 or L3-40) or placebo, followed by escalating gluten consumption (50 mg/d for 12 weeks, 1 g intermittent twice weekly for 12 weeks, 2 g/d sustained for 6 weeks, liberal diet for 1 year). RESULTS: Successful study completion rates at week 42 (primary outcome) were similar in each group (placebo: 57%, L3-20: 37%, and L3-40: 44%; P = 0.61), however gluten-related adverse events were significantly reduced in hookworm-treated participants: Median (range) adverse events/participant were as follows: placebo, 4 (1-9); L3-20, 1 (0-9); and L3-40, 0 (0-3) (P = 0.019). Duodenal villous height:crypt depth deteriorated similarly compared with their enrolment values in each group (mean change [95% confidence interval]: placebo, -0.6 [-1.3 to 0.2]; L3-20, -0.5 [-0.8 to 0.2]; and L3-40, -1.1 [-1.8 to 0.4]; P = 0.12). A retrospective analysis revealed that 9 of the 40 L3-treated participants failed to establish hookworm infections. Although week 42 completion rates were similar in hookworm-positive vs hookworm-negative participants (48% vs 44%, P = 0.43), quality of life symptom scores were lower in hookworm-positive participants after intermittent gluten challenge (mean [95% confidence interval]: 38.9 [33.9-44] vs 45.9 [39.2-52.6]). DISCUSSION: Hookworm infection does not restore tolerance to sustained moderate consumption of gluten (2 g/d) but was associated with improved symptom scores after intermittent consumption of lower, intermittent gluten doses.


Assuntos
Doença Celíaca/terapia , Glutens/imunologia , Larva/metabolismo , Necator americanus/metabolismo , Terapia com Helmintos/métodos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Doença Celíaca/imunologia , Método Duplo-Cego , Feminino , Glutens/administração & dosagem , Glutens/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Terapia com Helmintos/efeitos adversos , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa