Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3412, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649356

RESUMEN

Postnatal development of the gastrointestinal tract involves the establishment of the commensal microbiota, the acquisition of immune tolerance via a balanced immune cell composition, and maturation of the intestinal epithelium. While studies have uncovered an interplay between the first two, less is known about the role of the maturing epithelium. Here we show that intestinal-epithelial intrinsic expression of lysine-specific demethylase 1A (LSD1) is necessary for the postnatal maturation of intestinal epithelium and maintenance of this developed state during adulthood. Using microbiota-depleted mice, we find plasma cells, innate lymphoid cells (ILCs), and a specific myeloid population to depend on LSD1-controlled epithelial maturation. We propose that LSD1 controls the expression of epithelial-derived chemokines, such as Cxcl16, and that this is a mode of action for this epithelial-immune cell interplay in local ILC2s but not ILC3s. Together, our findings suggest that the maturing epithelium plays a dominant role in regulating the local immune cell composition, thereby contributing to gut homeostasis.


Asunto(s)
Microbioma Gastrointestinal , Histona Demetilasas , Mucosa Intestinal , Intestino Delgado , Animales , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Ratones , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Microbioma Gastrointestinal/inmunología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Ratones Endogámicos C57BL , Inmunidad Innata , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones Noqueados , Femenino , Masculino , Homeostasis
2.
Matrix Biol ; 127: 23-37, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38331051

RESUMEN

BACKGROUND: The kidney contains distinct glomerular and tubulointerstitial compartments with diverse cell types and extracellular matrix components. The role of immune cells in glomerular environment is crucial for dampening inflammation and maintaining homeostasis. Macrophages are innate immune cells that are influenced by their tissue microenvironment. However, the multifunctional role of kidney macrophages remains unclear. METHODS: Flow and imaging cytometry were used to determine the relative expression of CD81 and CX3CR1 (C-X3-C motif chemokine receptor 1) in kidney macrophages. Monocyte replenishment was assessed in Cx3cr1CreER X R26-yfp-reporter and shielded chimeric mice. Bulk RNA-sequencing and mass spectrometry-based proteomics were performed on isolated kidney macrophages from wild type and Col4a5-/- (Alport) mice. RNAscope was used to visualize transcripts and macrophage purity in bulk RNA assessed by CIBERSORTx analyses. RESULTS: In wild type mice we identified three distinct kidney macrophage subsets using CD81 and CX3CR1 and these subsets showed dependence on monocyte replenishment. In addition to their immune function, bulk RNA-sequencing of macrophages showed enrichment of biological processes associated with extracellular matrix. Proteomics identified collagen IV and laminins in kidney macrophages from wild type mice whilst other extracellular matrix proteins including cathepsins, ANXA2 and LAMP2 were enriched in Col4a5-/- (Alport) mice. A subset of kidney macrophages co-expressed matrix and macrophage transcripts. CONCLUSIONS: We identified CD81 and CX3CR1 positive kidney macrophage subsets with distinct dependence for monocyte replenishment. Multiomic analysis demonstrated that these cells have diverse functions that underscore the importance of macrophages in kidney health and disease.


Asunto(s)
Enfermedades Renales , Macrófagos , Ratones , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Macrófagos/metabolismo , Riñón/metabolismo , Inflamación/metabolismo , Enfermedades Renales/metabolismo , ARN/metabolismo
3.
Cerebrovasc Dis ; 51(4): 461-472, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34983048

RESUMEN

INTRODUCTION: Stroke is characterized by deleterious oxidative stress. Selenoprotein enzymes are essential endogenous antioxidants, and detailed insight into their role after stroke could define new therapeutic treatments. This systematic review aimed to elucidate how blood selenoprotein concentration and activity change in the acute phase of stroke. METHODS: We searched PubMed, EMBASE, and Medline databases for studies measuring serial blood selenoprotein concentration or activity in acute stroke patients or in stroke patients compared to non-stroke controls. Meta-analyses of studies stratified by the type of stroke, blood compartment, and type of selenoprotein measurement were conducted. RESULTS: Eighteen studies and data from 941 stroke patients and 708 non-stroke controls were included in this review. Glutathione peroxidase (GPx) was the only identified selenoprotein, and its activity was most frequently measured. Results from 12 studies and 693 patients showed that compared to non-stroke controls in acute ischaemic stroke patients, the GPx activity increased in haemolysate (standardized mean difference [SMD]: 0.27, 95% CI: 0.07-0.47) but decreased in plasma (mean difference [MD]: -1.08 U/L, 95% CI: -1.94 to -0.22) and serum (SMD: -0.54, 95% CI: -0.91 to -0.17). From 4 identified studies in 106 acute haemorrhagic stroke patients, the GPx activity decreased in haemolysate (SMD: -0.40, 95% CI: -0.68 to -0.13) and remained unchanged in plasma (MD: -0.10 U/L, 95% CI: -0.81 to 0.61) and serum (MD: -5.00 U/mL, 95% CI: -36.17 to 26.17) compared to non-stroke controls. Results from studies assessing the GPx activity in the haemolysate compartment were inconsistent and characterized by high heterogeneity. CONCLUSIONS: Our results suggest a reduction of the blood GPx activity in acute ischaemic stroke patients, a lack of evidence regarding a role for GPx in haemorrhagic stroke patients, and insufficient evidence for other selenoproteins.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Hemorrágico , Accidente Cerebrovascular Isquémico , Selenoproteínas , Antioxidantes , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/patología , Glutatión Peroxidasa , Accidente Cerebrovascular Hemorrágico/diagnóstico , Accidente Cerebrovascular Hemorrágico/patología , Humanos , Accidente Cerebrovascular Isquémico/diagnóstico , Accidente Cerebrovascular Isquémico/patología , Selenio , Selenoproteínas/metabolismo
4.
Immunology ; 164(4): 737-753, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34407221

RESUMEN

Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T-cell accumulation within the brain. Whilst the dynamics of CD8+ T-cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite-specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT-I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA-OVA infection. Whereas memory OT-I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT-I cells within the brain post-PbA-OVA infection displayed an enriched CD69+ CD103- profile and expressed low levels of T-bet. OT-I cells within the brain were excluded from short-term intravascular antibody labelling but were targeted effectively by longer-term systemically administered antibodies. Thus, the memory OT-I cells were extravascular within the brain post-ECM but were potentially not resident memory cells. Importantly, whilst memory OT-I cells exhibited strong reactivation during secondary PbA-OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA-OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post-ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T-cell populations within the brain and other tissues during repeat Plasmodium infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interacciones Huésped-Parásitos/inmunología , Malaria/inmunología , Malaria/parasitología , Plasmodium berghei/fisiología , Animales , Biomarcadores , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Quimiotaxis de Leucocito/inmunología , Susceptibilidad a Enfermedades , Epítopos de Linfocito T/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Matriz Extracelular , Memoria Inmunológica , Inmunofenotipificación , Estadios del Ciclo de Vida , Activación de Linfocitos/inmunología , Malaria/metabolismo , Malaria/patología , Malaria Cerebral/inmunología , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Ratones , Ratones Transgénicos , Especificidad de Órganos/inmunología
5.
Front Immunol ; 12: 715209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386014

RESUMEN

Peritoneal dialysis (PD) is a more continuous alternative to haemodialysis, for patients with chronic kidney disease, with considerable initial benefits for survival, patient independence and healthcare costs. However, long-term PD is associated with significant pathology, negating the positive effects over haemodialysis. Importantly, peritonitis and activation of macrophages is closely associated with disease progression and treatment failure. However, recent advances in macrophage biology suggest opposite functions for macrophages of different cellular origins. While monocyte-derived macrophages promote disease progression in some models of fibrosis, tissue resident macrophages have rather been associated with protective roles. Thus, we aimed to identify the relative contribution of tissue resident macrophages to PD induced inflammation in mice. Unexpectedly, we found an incremental loss of homeostatic characteristics, anti-inflammatory and efferocytic functionality in peritoneal resident macrophages, accompanied by enhanced inflammatory responses to external stimuli. Moreover, presence of glucose degradation products within the dialysis fluid led to markedly enhanced inflammation and almost complete disappearance of tissue resident cells. Thus, alterations in tissue resident macrophages may render long-term PD patients sensitive to developing peritonitis and consequently fibrosis/sclerosis.


Asunto(s)
Soluciones para Diálisis , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Diálisis Peritoneal , Animales , Plasticidad de la Célula , Femenino , Fibrosis , Glucosa/metabolismo , Inmunofenotipificación , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Diálisis Peritoneal/efectos adversos , Diálisis Peritoneal/métodos , Fenotipo
6.
Med ; 2(6): 720-735.e4, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33821250

RESUMEN

BACKGROUND: Emerging studies indicate that some coronavirus disease 2019 (COVID-19) patients suffer from persistent symptoms, including breathlessness and chronic fatigue; however, the long-term immune response in these patients presently remains ill-defined. METHODS: Here, we describe the phenotypic and functional characteristics of B and T cells in hospitalized COVID-19 patients during acute disease and at 3-6 months of convalescence. FINDINGS: We report that the alterations in B cell subsets observed in acute COVID-19 patients were largely recovered in convalescent patients. In contrast, T cells from convalescent patients displayed continued alterations with persistence of a cytotoxic program evident in CD8+ T cells as well as elevated production of type 1 cytokines and interleukin-17 (IL-17). Interestingly, B cells from patients with acute COVID-19 displayed an IL-6/IL-10 cytokine imbalance in response to Toll-like receptor activation, skewed toward a pro-inflammatory phenotype. Whereas the frequency of IL-6+ B cells was restored in convalescent patients irrespective of clinical outcome, the recovery of IL-10+ B cells was associated with the resolution of lung pathology. CONCLUSIONS: Our data detail lymphocyte alterations in previously hospitalized COVID-19 patients up to 6 months following hospital discharge and identify 3 subgroups of convalescent patients based on distinct lymphocyte phenotypes, with 1 subgroup associated with poorer clinical outcome. We propose that alterations in B and T cell function following hospitalization with COVID-19 could affect longer-term immunity and contribute to some persistent symptoms observed in convalescent COVID-19 patients. FUNDING: Provided by UKRI, Lister Institute of Preventative Medicine, the Wellcome Trust, The Kennedy Trust for Rheumatology Research, and 3M Global Giving.


Asunto(s)
COVID-19 , Linfocitos T CD8-positivos , Citocinas , Humanos , Interleucina-10 , Interleucina-6 , SARS-CoV-2
7.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33635312

RESUMEN

Hematopoietic stem cells reside in the bone marrow, where they generate the effector cells that drive immune responses. However, in response to inflammation, some hematopoietic stem and progenitor cells (HSPCs) are recruited to tissue sites and undergo extramedullary hematopoiesis. Contrasting with this paradigm, here we show residence and differentiation of HSPCs in healthy gingiva, a key oral barrier in the absence of overt inflammation. We initially defined a population of gingiva monocytes that could be locally maintained; we subsequently identified not only monocyte progenitors but also diverse HSPCs within the gingiva that could give rise to multiple myeloid lineages. Gingiva HSPCs possessed similar differentiation potentials, reconstitution capabilities, and heterogeneity to bone marrow HSPCs. However, gingival HSPCs responded differently to inflammatory insults, responding to oral but not systemic inflammation. Combined, we highlight a novel pathway of myeloid cell development at a healthy barrier, defining a gingiva-specific HSPC network that supports generation of a proportion of the innate immune cells that police this barrier.


Asunto(s)
Encía/citología , Encía/inmunología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Animales , Médula Ósea/metabolismo , Femenino , Hematopoyesis , Masculino , Ratones , Ratones Endogámicos C57BL , Mucosa Bucal/citología , Mucosa Bucal/inmunología , RNA-Seq/métodos , Análisis de la Célula Individual/métodos
8.
Sci Immunol ; 5(51)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943497

RESUMEN

COVID-19 pathogenesis is associated with an exaggerated immune response. However, the specific cellular mediators and inflammatory components driving diverse clinical disease outcomes remain poorly understood. We undertook longitudinal immune profiling on both whole blood and peripheral blood mononuclear cells (PBMCs) of hospitalized patients during the peak of the COVID-19 pandemic in the UK. Here, we report key immune signatures present shortly after hospital admission that were associated with the severity of COVID-19. Immune signatures were related to shifts in neutrophil to T cell ratio, elevated serum IL-6, MCP-1 and IP-10, and most strikingly, modulation of CD14+ monocyte phenotype and function. Modified features of CD14+ monocytes included poor induction of the prostaglandin-producing enzyme, COX-2, as well as enhanced expression of the cell cycle marker Ki-67. Longitudinal analysis revealed reversion of some immune features back to the healthy median level in patients with a good eventual outcome. These findings identify previously unappreciated alterations in the innate immune compartment of COVID-19 patients and lend support to the idea that therapeutic strategies targeting release of myeloid cells from bone marrow should be considered in this disease. Moreover, they demonstrate that features of an exaggerated immune response are present early after hospital admission suggesting immune-modulating therapies would be most beneficial at early timepoints.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Inmunidad Innata , Monocitos/inmunología , Neumonía Viral/inmunología , Adulto , Anciano , Biomarcadores/sangre , COVID-19 , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Ciclooxigenasa 2/inmunología , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Femenino , Interacciones Microbiota-Huesped/inmunología , Humanos , Mediadores de Inflamación/sangre , Mediadores de Inflamación/inmunología , Antígeno Ki-67/inmunología , Antígeno Ki-67/metabolismo , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Pandemias , Neumonía Viral/sangre , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Estudios Prospectivos , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Reino Unido/epidemiología
9.
J Exp Med ; 216(12): 2714-2723, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31582416

RESUMEN

Host immunity to parasitic nematodes requires the generation of a robust type 2 cytokine response, characterized by the production of interleukin 13 (IL-13), which drives expulsion. Here, we show that infection with helminths in the intestine also induces an ILC2-driven, IL-13-dependent goblet cell hyperplasia and increased production of mucins (Muc5b and Muc5ac) at distal sites, including the lungs and other mucosal barrier sites. Critically, we show that type 2 priming of lung tissue through increased mucin production inhibits the progression of a subsequent lung migratory helminth infection and limits its transit through the airways. These data show that infection by gastrointestinal-dwelling helminths induces a systemic innate mucin response that primes peripheral barrier sites for protection against subsequent secondary helminth infections. These data suggest that innate-driven priming of mucus barriers may have evolved to protect from subsequent infections with multiple helminth species, which occur naturally in endemic areas.


Asunto(s)
Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Moco/metabolismo , Animales , Protección Cruzada/inmunología , Células Caliciformes/citología , Células Caliciformes/metabolismo , Hiperplasia , Interleucina-13/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Masculino , Ratones , Ratones Noqueados , Mucinas/biosíntesis , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Triquinelosis/parasitología
10.
Front Immunol ; 10: 248, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30846985

RESUMEN

Cerebral malaria (CM) is one of the most severe complications of Plasmodium falciparum infection. There is evidence that repeated parasite exposure promotes resistance against CM. However, the immunological basis of this infection-induced resistance remains poorly understood. Here, utilizing the Plasmodium berghei ANKA (PbA) model of experimental cerebral malaria (ECM), we show that three rounds of infection and drug-cure protects against the development of ECM during a subsequent fourth (4X) infection. Exposure-induced resistance was associated with specific suppression of CD8+ T cell activation and CTL-related pathways, which corresponded with the development of heterogeneous atypical B cell populations as well as the gradual infection-induced generation and maintenance of high levels of anti-parasite IgG. Mechanistically, transfer of high-titer anti-parasite IgG did not protect 1X infected mice against ECM and depletion of atypical and regulatory B cells during 4X infection failed to abrogate infection-induced resistance to ECM. However, IgMi mice that were unable to produce secreted antibody, or undergo class switching, during the repeated rounds of infection failed to develop resistance against ECM. The failure of infection-induced protection in IgMi mice was associated with impaired development of atypical B cell populations and the inability to suppress pathogenic CD8+ T cell responses. Our results, therefore, suggest the importance of anti-parasite antibody responses, gradually acquired, and maintained through repeated Plasmodium infections, for modulating the B cell compartment and eventually suppressing memory CD8+ T cell reactivation to establish infection-induced resistance to ECM.


Asunto(s)
Formación de Anticuerpos/inmunología , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Animales , Encéfalo/parasitología , Linfocitos T CD8-positivos/parasitología , Activación de Linfocitos/inmunología , Malaria Cerebral/parasitología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos C57BL
11.
Sci Transl Med ; 10(464)2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30355800

RESUMEN

Macrophages in the healthy intestine are highly specialized and usually respond to the gut microbiota without provoking an inflammatory response. A breakdown in this tolerance leads to inflammatory bowel disease (IBD), but the mechanisms by which intestinal macrophages normally become conditioned to promote microbial tolerance are unclear. Strong epidemiological evidence linking disruption of the gut microbiota by antibiotic use early in life to IBD indicates an important role for the gut microbiota in modulating intestinal immunity. Here, we show that antibiotic use causes intestinal macrophages to become hyperresponsive to bacterial stimulation, producing excess inflammatory cytokines. Re-exposure of antibiotic-treated mice to conventional microbiota induced a long-term, macrophage-dependent increase in inflammatory T helper 1 (TH1) responses in the colon and sustained dysbiosis. The consequences of this dysregulated macrophage activity for T cell function were demonstrated by increased susceptibility to infections requiring TH17 and TH2 responses for clearance (bacterial Citrobacter rodentium and helminth Trichuris muris infections), corresponding with increased inflammation. Short-chain fatty acids (SCFAs) were depleted during antibiotic administration; supplementation of antibiotics with the SCFA butyrate restored the characteristic hyporesponsiveness of intestinal macrophages and prevented T cell dysfunction. Butyrate altered the metabolic behavior of macrophages to increase oxidative phosphorylation and also promoted alternative macrophage activation. In summary, the gut microbiota is essential to maintain macrophage-dependent intestinal immune homeostasis, mediated by SCFA-dependent pathways. Oral antibiotics disrupt this process to promote sustained T cell-mediated dysfunction and increased susceptibility to infections, highlighting important implications of repeated broad-spectrum antibiotic use.


Asunto(s)
Antibacterianos/farmacología , Homeostasis/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Intestinos/citología , Macrófagos/metabolismo , Linfocitos T/inmunología , Animales , Butiratos/farmacología , Citocinas/metabolismo , Ácidos Grasos/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/patología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Receptores CCR2/metabolismo , Linfocitos T/efectos de los fármacos , Células TH1/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 115(28): 7404-7409, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29954866

RESUMEN

Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1ß production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1ß release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.


Asunto(s)
Antimaláricos/farmacología , Encéfalo/parasitología , Sistemas de Liberación de Medicamentos/métodos , Interleucina-33/metabolismo , Malaria Cerebral/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Plasmodium falciparum/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Hemoproteínas/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-33/antagonistas & inhibidores , Macrófagos/metabolismo , Macrófagos/patología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Malaria Falciparum/metabolismo , Malaria Falciparum/patología , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Transcriptoma/efectos de los fármacos
13.
J Exp Med ; 215(6): 1507-1518, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29789388

RESUMEN

A defining feature of resident gut macrophages is their high replenishment rate from blood monocytes attributed to tonic commensal stimulation of this site. In contrast, almost all other tissues contain locally maintained macrophage populations, which coexist with monocyte-replenished cells at homeostasis. In this study, we identified three transcriptionally distinct mouse gut macrophage subsets that segregate based on expression of Tim-4 and CD4. Challenging current understanding, Tim-4+CD4+ gut macrophages were found to be locally maintained, while Tim-4-CD4+ macrophages had a slow turnover from blood monocytes; indeed, Tim-4-CD4- macrophages were the only subset with the high monocyte-replenishment rate currently attributed to gut macrophages. Moreover, all macrophage subpopulations required live microbiota to sustain their numbers, not only those derived from blood monocytes. These findings oppose the prevailing paradigm that all macrophages in the adult mouse gut rapidly turn over from monocytes in a microbiome-dependent manner; instead, these findings supplant it with a model of ontogenetic diversity where locally maintained subsets coexist with rapidly replaced monocyte-derived populations.


Asunto(s)
Antígenos CD4/metabolismo , Intestinos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Animales Recién Nacidos , Intestinos/microbiología , Ratones Endogámicos C57BL , Microbiota , Monocitos/metabolismo , Fenotipo , Receptores CCR2/metabolismo , Transcripción Genética
14.
Infect Immun ; 85(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28874445

RESUMEN

Experimental cerebral malaria (ECM) is a gamma interferon (IFN-γ)-dependent syndrome. However, whether IFN-γ promotes ECM through direct and synergistic targeting of multiple cell populations or by acting primarily on a specific responsive cell type is currently unknown. Here, using a panel of cell- and compartment-specific IFN-γ receptor 2 (IFN-γR2)-deficient mice, we show that IFN-γ causes ECM by signaling within both the hematopoietic and nonhematopoietic compartments. Mechanistically, hematopoietic and nonhematopoietic compartment-specific IFN-γR signaling exerts additive effects in orchestrating intracerebral inflammation, leading to the development of ECM. Surprisingly, mice with specific deletion of IFN-γR2 expression on myeloid cells, T cells, or neurons were completely susceptible to terminal ECM. Utilizing a reductionist in vitro system, we show that synergistic IFN-γ and tumor necrosis factor (TNF) stimulation promotes strong activation of brain blood vessel endothelial cells. Combined, our data show that within the hematopoietic compartment, IFN-γ causes ECM by acting redundantly or by targeting non-T cell or non-myeloid cell populations. Within the nonhematopoietic compartment, brain endothelial cells, but not neurons, may be the major target of IFN-γ leading to ECM development. Collectively, our data provide information on how IFN-γ mediates the development of cerebral pathology during malaria infection.


Asunto(s)
Encéfalo/inmunología , Células Endoteliales/inmunología , Interferón gamma/genética , Malaria Cerebral/genética , Plasmodium berghei/patogenicidad , Receptores de Interferón/genética , Animales , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Encéfalo/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/parasitología , Regulación de la Expresión Génica , Interferón gamma/inmunología , Interleucinas/genética , Interleucinas/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/parasitología , Neuronas/inmunología , Neuronas/parasitología , Plasmodium berghei/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/inmunología , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/parasitología
15.
PLoS Pathog ; 13(3): e1006267, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28273147

RESUMEN

The murine model of experimental cerebral malaria (ECM) has been utilised extensively in recent years to study the pathogenesis of human cerebral malaria (HCM). However, it has been proposed that the aetiologies of ECM and HCM are distinct, and, consequently, no useful mechanistic insights into the pathogenesis of HCM can be obtained from studying the ECM model. Therefore, in order to determine the similarities and differences in the pathology of ECM and HCM, we have performed the first spatial and quantitative histopathological assessment of the ECM syndrome. We demonstrate that the accumulation of parasitised red blood cells (pRBCs) in brain capillaries is a specific feature of ECM that is not observed during mild murine malaria infections. Critically, we show that individual pRBCs appear to occlude murine brain capillaries during ECM. As pRBC-mediated congestion of brain microvessels is a hallmark of HCM, this suggests that the impact of parasite accumulation on cerebral blood flow may ultimately be similar in mice and humans during ECM and HCM, respectively. Additionally, we demonstrate that cerebrovascular CD8+ T-cells appear to co-localise with accumulated pRBCs, an event that corresponds with development of widespread vascular leakage. As in HCM, we show that vascular leakage is not dependent on extensive vascular destruction. Instead, we show that vascular leakage is associated with alterations in transcellular and paracellular transport mechanisms. Finally, as in HCM, we observed axonal injury and demyelination in ECM adjacent to diverse vasculopathies. Collectively, our data therefore shows that, despite very different presentation, and apparently distinct mechanisms, of parasite accumulation, there appear to be a number of comparable features of cerebral pathology in mice and in humans during ECM and HCM, respectively. Thus, when used appropriately, the ECM model may be useful for studying specific pathological features of HCM.


Asunto(s)
Encéfalo/patología , Encéfalo/parasitología , Modelos Animales de Enfermedad , Malaria Cerebral/patología , Malaria Cerebral/parasitología , Animales , Eritrocitos/parasitología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Plasmodium berghei
16.
Pflugers Arch ; 469(3-4): 527-539, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28283748

RESUMEN

Monocyte-derived mononuclear phagocytes, particularly macrophages, are crucial to maintain gastrointestinal homeostasis in the steady state but are also important for protection against certain pathogens. However, when uncontrolled, they can promote immunopathology. Broadly two subsets of macrophages can be considered to perform the vast array of functions to complete these complex tasks: resident macrophages that dominate in the healthy gut and inflammation-elicited (inflammatory) macrophages that derive from circulating monocytes infiltrating inflamed tissue. Here, we discuss the features of resident and inflammatory intestinal macrophages, complexities in identifying and defining these populations and the mechanisms involved in their differentiation. In particular, focus will be placed on describing their unique ontogeny as well as local gastrointestinal signals that instruct specialisation of resident macrophages in healthy tissue. We then explore the very different roles of inflammatory macrophages and describe new data suggesting that they may be educated not only by the gut microenvironment but also by signals they receive during development in the bone marrow. Given the high degree of plasticity of gut macrophages and their multifaceted roles in both healthy and inflamed tissue, understanding the mechanisms controlling their differentiation could inform development of improved therapies for inflammatory diseases such as inflammatory bowel disease (IBD).


Asunto(s)
Homeostasis/fisiología , Inflamación/patología , Intestinos/patología , Intestinos/fisiología , Macrófagos/fisiología , Animales , Diferenciación Celular/fisiología , Humanos , Enfermedades Inflamatorias del Intestino/patología
17.
J Immunol ; 197(8): 3152-3164, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27630165

RESUMEN

CD4+ T cells that produce IFN-γ are the source of host-protective IL-10 during primary infection with a number of different pathogens, including Plasmodium spp. The fate of these CD4+IFN-γ+IL-10+ T cells following clearance of primary infection and their subsequent influence on the course of repeated infections is, however, presently unknown. In this study, utilizing IFN-γ-yellow fluorescent protein (YFP) and IL-10-GFP dual reporter mice, we show that primary malaria infection-induced CD4+YFP+GFP+ T cells have limited memory potential, do not stably express IL-10, and are disproportionately lost from the Ag-experienced CD4+ T cell memory population during the maintenance phase postinfection. CD4+YFP+GFP+ T cells generally exhibited a short-lived effector rather than effector memory T cell phenotype postinfection and expressed high levels of PD-1, Lag-3, and TIGIT, indicative of cellular exhaustion. Consistently, the surviving CD4+YFP+GFP+ T cell-derived cells were unresponsive and failed to proliferate during the early phase of secondary infection. In contrast, CD4+YFP+GFP- T cell-derived cells expanded rapidly and upregulated IL-10 expression during secondary infection. Correspondingly, CD4+ T cells were the major producers within an accelerated and amplified IL-10 response during the early stage of secondary malaria infection. Notably, IL-10 exerted quantitatively stronger regulatory effects on innate and CD4+ T cell responses during primary and secondary infections, respectively. The results in this study significantly improve our understanding of the durability of IL-10-producing CD4+ T cells postinfection and provide information on how IL-10 may contribute to optimized parasite control and prevention of immune-mediated pathology during repeated malaria infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Interleucina-10/metabolismo , Malaria/inmunología , Plasmodium/inmunología , Animales , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD4-Positivos/trasplante , Supervivencia Celular , Células Cultivadas , Humanos , Inmunosenescencia , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
18.
Infect Immun ; 84(1): 34-46, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26459508

RESUMEN

Immune-mediated pathology in interleukin-10 (IL-10)-deficient mice during blood-stage malaria infection typically manifests in nonlymphoid organs, such as the liver and lung. Thus, it is critical to define the cellular sources of IL-10 in these sensitive nonlymphoid compartments during infection. Moreover, it is important to determine if IL-10 production is controlled through conserved or disparate molecular programs in distinct anatomical locations during malaria infection, as this may enable spatiotemporal tuning of the regulatory immune response. In this study, using dual gamma interferon (IFN-γ)-yellow fluorescent protein (YFP) and IL-10-green fluorescent protein (GFP) reporter mice, we show that CD4(+) YFP(+) T cells are the major source of IL-10 in both lymphoid and nonlymphoid compartments throughout the course of blood-stage Plasmodium yoelii infection. Mature splenic CD4(+) YFP(+) GFP(+) T cells, which preferentially expressed high levels of CCR5, were capable of migrating to and seeding the nonlymphoid tissues, indicating that the systemically distributed host-protective cells have a common developmental history. Despite exhibiting comparable phenotypes, CD4(+) YFP(+) GFP(+) T cells from the liver and lung produced significantly larger quantities of IL-10 than their splenic counterparts, showing that the CD4(+) YFP(+) GFP(+) T cells exert graded functions in distinct tissue locations during infection. Unexpectedly, given the unique environmental conditions within discrete nonlymphoid and lymphoid organs, we show that IL-10 production by CD4(+) YFP(+) T cells is controlled systemically during malaria infection through IL-27 receptor signaling that is supported after CD4(+) T cell priming by ICOS signaling. The results in this study substantially improve our understanding of the systemic IL-10 response to malaria infection, particularly within sensitive nonlymphoid organs.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-10/inmunología , Interleucinas/inmunología , Malaria/inmunología , Traslado Adoptivo , Animales , Proteínas Bacterianas/genética , Linfocitos T CD4-Positivos/trasplante , Proteínas Fluorescentes Verdes/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Hígado/inmunología , Proteínas Luminiscentes/genética , Pulmón/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasmodium yoelii/inmunología , Bazo/inmunología
19.
PLoS Pathog ; 11(11): e1005210, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26562533

RESUMEN

There is significant evidence that brain-infiltrating CD8+ T cells play a central role in the development of experimental cerebral malaria (ECM) during Plasmodium berghei ANKA infection of C57BL/6 mice. However, the mechanisms through which they mediate their pathogenic activity during malaria infection remain poorly understood. Utilizing intravital two-photon microscopy combined with detailed ex vivo flow cytometric analysis, we show that brain-infiltrating T cells accumulate within the perivascular spaces of brains of mice infected with both ECM-inducing (P. berghei ANKA) and non-inducing (P. berghei NK65) infections. However, perivascular T cells displayed an arrested behavior specifically during P. berghei ANKA infection, despite the brain-accumulating CD8+ T cells exhibiting comparable activation phenotypes during both infections. We observed T cells forming long-term cognate interactions with CX3CR1-bearing antigen presenting cells within the brains during P. berghei ANKA infection, but abrogation of this interaction by targeted depletion of the APC cells failed to prevent ECM development. Pathogenic CD8+ T cells were found to colocalize with rare apoptotic cells expressing CD31, a marker of endothelial cells, within the brain during ECM. However, cellular apoptosis was a rare event and did not result in loss of cerebral vasculature or correspond with the extensive disruption to its integrity observed during ECM. In summary, our data show that the arrest of T cells in the perivascular compartments of the brain is a unique signature of ECM-inducing malaria infection and implies an important role for this event in the development of the ECM-syndrome.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Malaria Cerebral/inmunología , Malaria Falciparum/microbiología , Parasitemia/inmunología , Plasmodium berghei/inmunología , Animales , Linfocitos T CD8-positivos/parasitología , Modelos Animales de Enfermedad , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones Endogámicos C57BL
20.
PLoS Pathog ; 11(2): e1004607, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25659095

RESUMEN

Cerebral malaria (CM) is a complex parasitic disease caused by Plasmodium sp. Failure to establish an appropriate balance between pro- and anti-inflammatory immune responses is believed to contribute to the development of cerebral pathology. Using the blood-stage PbA (Plasmodium berghei ANKA) model of infection, we show here that administration of the pro-Th2 cytokine, IL-33, prevents the development of experimental cerebral malaria (ECM) in C57BL/6 mice and reduces the production of inflammatory mediators IFN-γ, IL-12 and TNF-α. IL-33 drives the expansion of type-2 innate lymphoid cells (ILC2) that produce Type-2 cytokines (IL-4, IL-5 and IL-13), leading to the polarization of the anti-inflammatory M2 macrophages, which in turn expand Foxp3 regulatory T cells (Tregs). PbA-infected mice adoptively transferred with ILC2 have elevated frequency of M2 and Tregs and are protected from ECM. Importantly, IL-33-treated mice deleted of Tregs (DEREG mice) are no longer able to resist ECM. Our data therefore provide evidence that IL-33 can prevent the development of ECM by orchestrating a protective immune response via ILC2, M2 macrophages and Tregs.


Asunto(s)
Interleucina-33/inmunología , Macrófagos/inmunología , Malaria Cerebral/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...