Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nat Rev Genet ; 25(4): 235, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38291235
2.
Mucosal Immunol ; 17(1): 137-146, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37967720

RESUMEN

The intestine is home to an intertwined network of epithelial, immune, and neuronal cells as well as the microbiome, with implications for immunity, systemic metabolism, and behavior. While the complexity of this microenvironment has long since been acknowledged, recent technological advances have propelled our understanding to an unprecedented level. Notably, the microbiota and non-immune or structural cells have emerged as important conductors of intestinal immunity, and by contrast, cells of both the innate and adaptive immune systems have demonstrated non-canonical roles in tissue repair and metabolism. This review highlights recent works in the following two streams: non-immune cells of the intestine performing immunological functions; and traditional immune cells exhibiting non-immune functions in the gut.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Inmunidad Innata , Mucosa Intestinal
3.
Nat Rev Gastroenterol Hepatol ; 20(9): 597-614, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37258747

RESUMEN

The number of studies investigating the human gastrointestinal tract using various single-cell profiling methods has increased substantially in the past few years. Although this increase provides a unique opportunity for the generation of the first comprehensive Human Gut Cell Atlas (HGCA), there remains a range of major challenges ahead. Above all, the ultimate success will largely depend on a structured and coordinated approach that aligns global efforts undertaken by a large number of research groups. In this Roadmap, we discuss a comprehensive forward-thinking direction for the generation of the HGCA on behalf of the Gut Biological Network of the Human Cell Atlas. Based on the consensus opinion of experts from across the globe, we outline the main requirements for the first complete HGCA by summarizing existing data sets and highlighting anatomical regions and/or tissues with limited coverage. We provide recommendations for future studies and discuss key methodologies and the importance of integrating the healthy gut atlas with related diseases and gut organoids. Importantly, we critically overview the computational tools available and provide recommendations to overcome key challenges.


Asunto(s)
Tracto Gastrointestinal , Organoides , Humanos , Predicción
4.
Blood ; 141(19): 2343-2358, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-36758207

RESUMEN

Classic Hodgkin lymphoma (cHL) has a rich immune infiltrate, which is an intrinsic component of the neoplastic process. Malignant Hodgkin Reed-Sternberg cells (HRSCs) create an immunosuppressive microenvironment by the expression of regulatory molecules, preventing T-cell activation. It has also been demonstrated that mononuclear phagocytes (MNPs) in the vicinity of HRSCs express similar regulatory mechanisms in parallel, and their presence in tissue is associated with inferior patient outcomes. MNPs in cHL have hitherto been identified by a small number of canonical markers and are usually described as tumor-associated macrophages. The organization of MNP networks and interactions with HRSCs remains unexplored at high resolution. Here, we defined the global immune-cell composition of cHL and nonlymphoma lymph nodes, integrating data across single-cell RNA sequencing, spatial transcriptomics, and multiplexed immunofluorescence. We observed that MNPs comprise multiple subsets of monocytes, macrophages, and dendritic cells (DCs). Classical monocytes, macrophages and conventional DC2s were enriched in the vicinity of HRSCs, but plasmacytoid DCs and activated DCs were excluded. Unexpectedly, cDCs and monocytes expressed immunoregulatory checkpoints PD-L1, TIM-3, and the tryptophan-catabolizing protein IDO, at the same level as macrophages. Expression of these molecules increased with age. We also found that classical monocytes are important signaling hubs, potentially controlling the retention of cDC2 and ThExh via CCR1-, CCR4-, CCR5-, and CXCR3-dependent signaling. Enrichment of the cDC2-monocyte-macrophage network in diagnostic biopsies is associated with early treatment failure. These results reveal unanticipated complexity and spatial polarization within the MNP compartment, further demonstrating their potential roles in immune evasion by cHL.


Asunto(s)
Enfermedad de Hodgkin , Humanos , Enfermedad de Hodgkin/diagnóstico , Células de Reed-Sternberg/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Inmunosupresores , Microambiente Tumoral
5.
Gastroenterology ; 162(3): 859-876, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34780721

RESUMEN

BACKGROUND & AIMS: Monogenic forms of inflammatory bowel disease (IBD) illustrate the essential roles of individual genes in pathways and networks safeguarding immune tolerance and gut homeostasis. METHODS: To build a taxonomy model, we assessed 165 disorders. Genes were prioritized based on penetrance of IBD and disease phenotypes were integrated with multi-omics datasets. Monogenic IBD genes were classified by (1) overlapping syndromic features, (2) response to hematopoietic stem cell transplantation, (3) bulk RNA-sequencing of 32 tissues, (4) single-cell RNA-sequencing of >50 cell subsets from the intestine of healthy individuals and patients with IBD (pediatric and adult), and (5) proteomes of 43 immune subsets. The model was validated by addition of newly identified monogenic IBD defects. As a proof-of-concept, we explore the intersection between immunometabolism and antimicrobial activity for a group of disorders (G6PC3/SLC37A4). RESULTS: Our quantitative integrated taxonomy defines the cellular landscape of monogenic IBD gene expression across 102 genes with high and moderate penetrance (81 in the model set and 21 genes in the validation set). We illustrate distinct cellular networks, highlight expression profiles across understudied cell types (e.g., CD8+ T cells, neutrophils, epithelial subsets, and endothelial cells) and define genotype-phenotype associations (perianal disease and defective antimicrobial activity). We illustrate processes and pathways shared across cellular compartments and phenotypic groups and highlight cellular immunometabolism with mammalian target of rapamycin activation as one of the converging pathways. There is an overlap of genes and enriched cell-specific expression between monogenic and polygenic IBD. CONCLUSION: Our taxonomy integrates genetic, clinical and multi-omic data; providing a basis for genomic diagnostics and testable hypotheses for disease functions and treatment responses.


Asunto(s)
Enfermedades Inflamatorias del Intestino/clasificación , Enfermedades Inflamatorias del Intestino/genética , Edad de Inicio , Antiportadores/genética , Células Cultivadas , Clasificación , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Genotipo , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfato/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Macrófagos , Metabolómica , Proteínas de Transporte de Monosacáridos/genética , Penetrancia , Fenotipo , Transducción de Señal/genética
6.
Nature ; 597(7875): 196-205, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497388

RESUMEN

The Human Developmental Cell Atlas (HDCA) initiative, which is part of the Human Cell Atlas, aims to create a comprehensive reference map of cells during development. This will be critical to understanding normal organogenesis, the effect of mutations, environmental factors and infectious agents on human development, congenital and childhood disorders, and the cellular basis of ageing, cancer and regenerative medicine. Here we outline the HDCA initiative and the challenges of mapping and modelling human development using state-of-the-art technologies to create a reference atlas across gestation. Similar to the Human Genome Project, the HDCA will integrate the output from a growing community of scientists who are mapping human development into a unified atlas. We describe the early milestones that have been achieved and the use of human stem-cell-derived cultures, organoids and animal models to inform the HDCA, especially for prenatal tissues that are hard to acquire. Finally, we provide a roadmap towards a complete atlas of human development.


Asunto(s)
Movimiento Celular , Rastreo Celular , Células/citología , Biología Evolutiva/métodos , Embrión de Mamíferos/citología , Feto/citología , Difusión de la Información , Organogénesis , Adulto , Animales , Atlas como Asunto , Técnicas de Cultivo de Célula , Supervivencia Celular , Visualización de Datos , Femenino , Humanos , Imagenología Tridimensional , Masculino , Modelos Animales , Organogénesis/genética , Organoides/citología , Células Madre/citología
7.
Nature ; 597(7875): 250-255, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497389

RESUMEN

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Asunto(s)
Envejecimiento , Sistema Nervioso Entérico/citología , Feto/citología , Salud , Intestinos/citología , Intestinos/crecimiento & desarrollo , Ganglios Linfáticos/citología , Ganglios Linfáticos/crecimiento & desarrollo , Adulto , Animales , Niño , Enfermedad de Crohn/patología , Conjuntos de Datos como Asunto , Sistema Nervioso Entérico/anatomía & histología , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/crecimiento & desarrollo , Células Epiteliales/citología , Femenino , Feto/anatomía & histología , Feto/embriología , Humanos , Intestinos/embriología , Intestinos/inervación , Ganglios Linfáticos/embriología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Organogénesis , Receptores de IgG/metabolismo , Transducción de Señal , Análisis Espacio-Temporal , Factores de Tiempo
8.
Dev Cell ; 55(6): 771-783.e5, 2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33290721

RESUMEN

Human gut development requires the orchestrated interaction of differentiating cell types. Here, we generate an in-depth single-cell map of the developing human intestine at 6-10 weeks post-conception. Our analysis reveals the transcriptional profile of cycling epithelial precursor cells; distinct from LGR5-expressing cells. We propose that these cells may contribute to differentiated cell subsets via the generation of LGR5-expressing stem cells and receive signals from surrounding mesenchymal cells. Furthermore, we draw parallels between the transcriptomes of ex vivo tissues and in vitro fetal organoids, revealing the maturation of organoid cultures in a dish. Lastly, we compare scRNA-seq profiles from pediatric Crohn's disease epithelium alongside matched healthy controls to reveal disease-associated changes in the epithelial composition. Contrasting these with the fetal profiles reveals the re-activation of fetal transcription factors in Crohn's disease. Our study provides a resource available at www.gutcellatlas.org, and underscores the importance of unraveling fetal development in understanding disease.


Asunto(s)
Enfermedad de Crohn/genética , Mucosa Intestinal/metabolismo , Transcriptoma , Adolescente , Células Cultivadas , Niño , Enfermedad de Crohn/metabolismo , Humanos , Mucosa Intestinal/embriología , RNA-Seq , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Nat Immunol ; 21(12): 1597-1610, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046889

RESUMEN

The dynamics of CD4+ T cell memory development remain to be examined at genome scale. In malaria-endemic regions, antimalarial chemoprevention protects long after its cessation and associates with effects on CD4+ T cells. We applied single-cell RNA sequencing and computational modelling to track memory development during Plasmodium infection and treatment. In the absence of central memory precursors, two trajectories developed as T helper 1 (TH1) and follicular helper T (TFH) transcriptomes contracted and partially coalesced over three weeks. Progeny of single clones populated TH1 and TFH trajectories, and fate-mapping suggested that there was minimal lineage plasticity. Relationships between TFH and central memory were revealed, with antimalarials modulating these responses and boosting TH1 recall. Finally, single-cell epigenomics confirmed that heterogeneity among effectors was partially reset in memory. Thus, the effector-to-memory transition in CD4+ T cells is gradual during malaria and is modulated by antiparasitic drugs. Graphical user interfaces are presented for examining gene-expression dynamics and gene-gene correlations ( http://haquelab.mdhs.unimelb.edu.au/cd4_memory/ ).


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Memoria Inmunológica , Malaria/inmunología , Plasmodium/inmunología , Transcriptoma , Traslado Adoptivo , Animales , Antimaláricos/farmacología , Biomarcadores , Cromatina/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Malaria/parasitología , Malaria/terapia , Ratones , Plasmodium/efectos de los fármacos
11.
Cell ; 181(6): 1291-1306.e19, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32407674

RESUMEN

Enteroendocrine cells (EECs) sense intestinal content and release hormones to regulate gastrointestinal activity, systemic metabolism, and food intake. Little is known about the molecular make-up of human EEC subtypes and the regulated secretion of individual hormones. Here, we describe an organoid-based platform for functional studies of human EECs. EEC formation is induced in vitro by transient expression of NEUROG3. A set of gut organoids was engineered in which the major hormones are fluorescently tagged. A single-cell mRNA atlas was generated for the different EEC subtypes, and their secreted products were recorded by mass-spectrometry. We note key differences to murine EECs, including hormones, sensory receptors, and transcription factors. Notably, several hormone-like molecules were identified. Inter-EEC communication is exemplified by secretin-induced GLP-1 secretion. Indeed, individual EEC subtypes carry receptors for various EEC hormones. This study provides a rich resource to study human EEC development and function.


Asunto(s)
Células Enteroendocrinas/metabolismo , ARN Mensajero/genética , Células Cultivadas , Hormonas Gastrointestinales/genética , Tracto Gastrointestinal/metabolismo , Péptido 1 Similar al Glucagón/genética , Humanos , Organoides/metabolismo , Factores de Transcripción/genética , Transcriptoma/genética
12.
Immunol Cell Biol ; 98(6): 428-430, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32418276

RESUMEN

Chen et al. describe how B-cell clones observed in the gut of many different individuals (recurrent or "public" clonotypes) are shaped by the combined influences of common microbial antigens and underlying genomic recombination biases.


Asunto(s)
Microbioma Gastrointestinal , Linfocitos B , Células Clonales , Células Germinativas , Centro Germinal , Humanos
13.
J Immunol ; 204(11): 2949-2960, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32321759

RESUMEN

Despite extensive mapping of long noncoding RNAs in immune cells, their function in vivo remains poorly understood. In this study, we identify over 100 long noncoding RNAs that are differentially expressed within 24 h of Th1 cell activation. Among those, we show that suppression of Malat1 is a hallmark of CD4+ T cell activation, but its complete deletion results in more potent immune responses to infection. This is because Malat1-/- Th1 and Th2 cells express lower levels of the immunosuppressive cytokine IL-10. In vivo, the reduced CD4+ T cell IL-10 expression in Malat1-/- mice underpins enhanced immunity and pathogen clearance in experimental visceral leishmaniasis (Leishmania donovani) but more severe disease in a model of malaria (Plasmodium chabaudi chabaudi AS). Mechanistically, Malat1 regulates IL-10 through enhancing expression of Maf, a key transcriptional regulator of IL-10 Maf expression correlates with Malat1 in single Ag-specific Th cells from P. chabaudi chabaudi AS-infected mice and is downregulated in Malat1-/- Th1 and Th2 cells. The Malat1 RNA is responsible for these effects, as antisense oligonucleotide-mediated inhibition of Malat1 also suppresses Maf and IL-10 levels. Our results reveal that through promoting expression of the Maf/IL-10 axis in effector Th cells, Malat1 is a nonredundant regulator of mammalian immunity.


Asunto(s)
Interleucina-10/metabolismo , Leishmania donovani/fisiología , Leishmaniasis Visceral/inmunología , Proteínas Proto-Oncogénicas c-maf/metabolismo , ARN Largo no Codificante/genética , Células TH1/inmunología , Células Th2/inmunología , Animales , Femenino , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica , Inmunidad/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-maf/genética , Regulación hacia Arriba
14.
Annu Rev Immunol ; 38: 727-757, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-32075461

RESUMEN

Immune cells are characterized by diversity, specificity, plasticity, and adaptability-properties that enable them to contribute to homeostasis and respond specifically and dynamically to the many threats encountered by the body. Single-cell technologies, including the assessment of transcriptomics, genomics, and proteomics at the level of individual cells, are ideally suited to studying these properties of immune cells. In this review we discuss the benefits of adopting single-cell approaches in studying underappreciated qualities of immune cells and highlight examples where these technologies have been critical to advancing our understanding of the immune system in health and disease.


Asunto(s)
Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad , Análisis de la Célula Individual , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Homeostasis , Humanos , Sistema Inmunológico/citología , Imagen Molecular , Análisis de la Célula Individual/métodos
15.
Nat Immunol ; 21(3): 343-353, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066951

RESUMEN

Gastrointestinal microbiota and immune cells interact closely and display regional specificity; however, little is known about how these communities differ with location. Here, we simultaneously assess microbiota and single immune cells across the healthy, adult human colon, with paired characterization of immune cells in the mesenteric lymph nodes, to delineate colonic immune niches at steady state. We describe distinct helper T cell activation and migration profiles along the colon and characterize the transcriptional adaptation trajectory of regulatory T cells between lymphoid tissue and colon. Finally, we show increasing B cell accumulation, clonal expansion and mutational frequency from the cecum to the sigmoid colon and link this to the increasing number of reactive bacterial species.


Asunto(s)
Colon/inmunología , Colon/microbiología , Microbioma Gastrointestinal/inmunología , Adulto , Linfocitos B/inmunología , Colon/citología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Especificidad de Órganos , RNA-Seq , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Transcriptoma
16.
PLoS Pathog ; 15(2): e1007599, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811498

RESUMEN

Plasmodium parasites invade and multiply inside red blood cells (RBC). Through a cycle of maturation, asexual replication, rupture and release of multiple infective merozoites, parasitised RBC (pRBC) can reach very high numbers in vivo, a process that correlates with disease severity in humans and experimental animals. Thus, controlling pRBC numbers can prevent or ameliorate malaria. In endemic regions, circulating parasite-specific antibodies associate with immunity to high parasitemia. Although in vitro assays reveal that protective antibodies could control pRBC via multiple mechanisms, in vivo assessment of antibody function remains challenging. Here, we employed two mouse models of antibody-mediated immunity to malaria, P. yoelii 17XNL and P. chabaudi chabaudi AS infection, to study infection-induced, parasite-specific antibody function in vivo. By tracking a single generation of pRBC, we tested the hypothesis that parasite-specific antibodies accelerate pRBC clearance. Though strongly protective against homologous re-challenge, parasite-specific IgG did not alter the rate of pRBC clearance, even in the presence of ongoing, systemic inflammation. Instead, antibodies prevented parasites progressing from one generation of RBC to the next. In vivo depletion studies using clodronate liposomes or cobra venom factor, suggested that optimal antibody function required splenic macrophages and dendritic cells, but not complement C3/C5-mediated killing. Finally, parasite-specific IgG bound poorly to the surface of pRBC, yet strongly to structures likely exposed by the rupture of mature schizonts. Thus, in our models of humoral immunity to malaria, infection-induced antibodies did not accelerate pRBC clearance, and instead co-operated with splenic phagocytes to block subsequent generations of pRBC.


Asunto(s)
Malaria/inmunología , Malaria/metabolismo , Plasmodium/crecimiento & desarrollo , Animales , Anticuerpos Antiprotozoarios/metabolismo , Modelos Animales de Enfermedad , Eritrocitos/microbiología , Eritrocitos/fisiología , Humanos , Ratones , Parásitos , Fagocitos , Plasmodium/metabolismo , Plasmodium/patogenicidad , Plasmodium chabaudi/inmunología , Plasmodium chabaudi/patogenicidad , Plasmodium yoelii/inmunología , Plasmodium yoelii/patogenicidad
17.
Front Immunol ; 9: 1435, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29997618

RESUMEN

The single-cell revolution is paving the way towards the molecular characterisation of every cell type in the human body, revealing relationships between cell types and states at high resolution. Changes in cellular phenotypes are particularly prevalent in the immune system and can be observed in its continuous remodelling up to adulthood, response to disease and development of immunological memory. In this review, we delve into the world of cellular dynamics of the immune system. We discuss current single-cell experimental and computational approaches in this area, giving insights into plasticity and commitment of cell fates. Finally, we provide an outlook on upcoming technological developments and predict how these will improve our understanding of the immune system.

18.
J Immunol ; 200(4): 1443-1456, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29321276

RESUMEN

Differentiation of CD4+ Th cells is critical for immunity to malaria. Several innate immune signaling pathways have been implicated in the detection of blood-stage Plasmodium parasites, yet their influence over Th cell immunity remains unclear. In this study, we used Plasmodium-reactive TCR transgenic CD4+ T cells, termed PbTII cells, during nonlethal P. chabaudi chabaudi AS and P. yoelii 17XNL infection in mice, to examine Th cell development in vivo. We found no role for caspase1/11, stimulator of IFN genes, or mitochondrial antiviral-signaling protein, and only modest roles for MyD88 and TRIF-dependent signaling in controlling PbTII cell expansion. In contrast, IFN regulatory factor 3 (IRF3) was important for supporting PbTII expansion, promoting Th1 over T follicular helper (Tfh) differentiation, and controlling parasites during the first week of infection. IRF3 was not required for early priming by conventional dendritic cells, but was essential for promoting CXCL9 and MHC class II expression by inflammatory monocytes that supported PbTII responses in the spleen. Thereafter, IRF3-deficiency boosted Tfh responses, germinal center B cell and memory B cell development, parasite-specific Ab production, and resolution of infection. We also noted a B cell-intrinsic role for IRF3 in regulating humoral immune responses. Thus, we revealed roles for IRF3 in balancing Th1- and Tfh-dependent immunity during nonlethal infection with blood-stage Plasmodium parasites.


Asunto(s)
Diferenciación Celular/inmunología , Factor 3 Regulador del Interferón/inmunología , Malaria/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/inmunología , Animales , Femenino , Centro Germinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Bazo/inmunología
19.
J Immunol ; 199(12): 4165-4179, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29084838

RESUMEN

We describe an MHC class II (I-Ab)-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/inmunología , Células Dendríticas/inmunología , Malaria/inmunología , Ratones Transgénicos/inmunología , Parasitemia/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Protozoos/inmunología , Antígenos CD40/deficiencia , Ligando de CD40/inmunología , Células Cultivadas , Cruzamientos Genéticos , Hibridomas , Activación de Linfocitos , Malaria Cerebral/inmunología , Malaria Cerebral/prevención & control , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos/genética , Plasmodium berghei/inmunología , Quimera por Radiación
20.
Int J Parasitol ; 47(14): 913-922, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28864033

RESUMEN

The artemisinins are the first-line therapy for severe and uncomplicated malaria, since they cause rapid declines in parasitemia after treatment. Despite this, in vivo mechanisms underlying this rapid decline remain poorly characterised. The overall decline in parasitemia is the net effect of drug inhibition of parasites and host clearance, which competes against any ongoing parasite proliferation. Separating these mechanisms in vivo was not possible through measurements of total parasitemia alone. Therefore, we employed an adoptive transfer approach in which C57BL/6J mice were transfused with Plasmodium berghei ANKA strain-infected, fluorescent red blood cells, and subsequently drug-treated. This approach allowed us to distinguish between the initial drug-treated generation of parasites (Gen0), and their progeny (Gen1). Artesunate efficiently impaired maturation of Gen0 parasites, such that a sufficiently high dose completely arrested maturation after 6h of in vivo exposure. In addition, artesunate-affected parasites were cleared from circulation with a half-life of 6.7h. In vivo cell depletion studies using clodronate liposomes revealed an important role for host phagocytes in the removal of artesunate-affected parasites, particularly ring and trophozoite stages. Finally, we found that a second antimalarial drug, mefloquine, was less effective than artesunate at suppressing parasite maturation and driving host-mediated parasite clearance. Thus, we propose that in vivo artesunate treatment causes rapid decline in parasitemia by arresting parasite maturation and encouraging phagocyte-mediated clearance of parasitised RBCs.


Asunto(s)
Antimaláricos/farmacología , Malaria/tratamiento farmacológico , Parasitemia/tratamiento farmacológico , Plasmodium berghei/efectos de los fármacos , Plasmodium yoelii/efectos de los fármacos , Traslado Adoptivo , Animales , Antimaláricos/administración & dosificación , Artemisininas/administración & dosificación , Artemisininas/farmacología , Artesunato , Relación Dosis-Respuesta a Droga , Eritrocitos/parasitología , Femenino , Citometría de Flujo , Malaria/parasitología , Mefloquina/administración & dosificación , Mefloquina/farmacología , Ratones , Ratones Endogámicos C57BL , Parasitemia/parasitología , Fagocitos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium yoelii/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA