Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
2.
Nature ; 570(7762): 528-532, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31168092

RESUMEN

Tuberculosis is the leading cause of death by an infectious disease worldwide1. However, the involvement of innate lymphoid cells (ILCs) in immune responses to infection with Mycobacterium tuberculosis (Mtb) is unknown. Here we show that circulating subsets of ILCs are depleted from the blood of participants with pulmonary tuberculosis and restored upon treatment. Tuberculosis increased accumulation of ILC subsets in the human lung, coinciding with a robust transcriptional response to infection, including a role in orchestrating the recruitment of immune subsets. Using mouse models, we show that group 3 ILCs (ILC3s) accumulated rapidly in Mtb-infected lungs and coincided with the accumulation of alveolar macrophages. Notably, mice that lacked ILC3s exhibited a reduction in the accumulation of early alveolar macrophages and decreased Mtb control. We show that the C-X-C motif chemokine receptor 5 (CXCR5)-C-X-C motif chemokine ligand 13 (CXCL13) axis is involved in Mtb control, as infection upregulates CXCR5 on circulating ILC3s and increases plasma levels of its ligand, CXCL13, in humans. Moreover, interleukin-23-dependent expansion of ILC3s in mice and production of interleukin-17 and interleukin-22 were found to be critical inducers of lung CXCL13, early innate immunity and the formation of protective lymphoid follicles within granulomas. Thus, we demonstrate an early protective role for ILC3s in immunity to Mtb infection.


Asunto(s)
Inmunidad Innata/inmunología , Linfocitos/clasificación , Linfocitos/inmunología , Macrófagos Alveolares/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Animales , Quimiocina CXCL13/inmunología , Femenino , Granuloma/inmunología , Granuloma/patología , Humanos , Interleucina-17/inmunología , Interleucinas/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Linfocitos/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Receptores CXCR5/inmunología , Transcriptoma/genética , Tuberculosis Pulmonar/genética , Interleucina-22
3.
Am J Respir Cell Mol Biol ; 64(3): 318-330, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33264084

RESUMEN

Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-ß-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvß3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Pulmón/irrigación sanguínea , Pulmón/crecimiento & desarrollo , FN-kappa B/metabolismo , Neovascularización Fisiológica , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Movimiento Celular , Factores Estimulantes de Colonias/metabolismo , Células Endoteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Ratones Endogámicos C57BL , Óxido Nítrico/biosíntesis , Nacimiento Prematuro , Alveolos Pulmonares/metabolismo , Ovinos
4.
Am J Respir Crit Care Med ; 193(2): 186-97, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26417909

RESUMEN

RATIONALE: Autologous and allogeneic hematopoietic stem cell transplant (HSCT) patients are susceptible to pulmonary infections, including bacterial pathogens, even after hematopoietic reconstitution. We previously reported that murine bone marrow transplant (BMT) neutrophils overexpress cyclooxygenase-2, overproduce prostaglandin E2 (PGE2), and exhibit defective intracellular bacterial killing. Neutrophil extracellular traps (NETs) are DNA structures that capture and kill extracellular bacteria and other pathogens. OBJECTIVES: To determine whether NETosis was defective after transplant and if so, whether this was regulated by PGE2 signaling. METHODS: Neutrophils isolated from mice and humans (both control and HSCT subjects) were analyzed for NETosis in response to various stimuli in the presence or absence of PGE2 signaling modifiers. MEASUREMENTS AND MAIN RESULTS: NETs were visualized by immunofluorescence or quantified by Sytox Green fluorescence. Treatment of BMT or HSCT neutrophils with phorbol 12-myristate 13-acetate or rapamycin resulted in reduced NET formation relative to control cells. NET formation after BMT was rescued both in vitro and in vivo with cyclooxygenase inhibitors. Additionally, the EP2 receptor antagonist (PF-04418948) or the EP4 antagonist (AE3-208) restored NET formation in neutrophils isolated from BMT mice or HSCT patients. Exogenous PGE2 treatment limited NETosis of neutrophils collected from normal human volunteers and naive mice in an exchange protein activated by cAMP- and protein kinase A-dependent manner. CONCLUSIONS: Our results suggest blockade of the PGE2-EP2 or EP4 signaling pathway restores NETosis after transplantation. Furthermore, these data provide the first description of a physiologic inhibitor of NETosis.


Asunto(s)
Dinoprostona/inmunología , Trampas Extracelulares/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Adulto , Anciano , Animales , Dinoprostona/farmacología , Trampas Extracelulares/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Adulto Joven
5.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L86-95, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361568

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is complicated by pulmonary infections that manifest posttransplantation. Despite engraftment, susceptibility to infections persists long after reconstitution. Previous work using a murine bone marrow transplant (BMT) model implicated increased cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) in promoting impaired alveolar macrophage (AM) responses. However, mechanisms driving COX-2 overexpression remained elusive. Previously, transforming growth factor-ß (TGF-ß) signaling after BMT was shown to promote hypomethylation of the COX-2 gene. Here, we provide mechanistic insight into how this occurs and show that TGF-ß induces microRNA (miR)-29b while decreasing DNA methyltransferases (DNMT)1, DNMT3a, and DNMT3b in AMs after BMT. De novo DNMT3a and DNMT3b were decreased upon transient transfection of miR-29b, resulting in decreased methylation of the COX-2 promoter and induction of COX-2. As a consequence, miR-29b-driven upregulation of COX-2 promoted AM dysfunction, and transfection of BMT AMs with a miR-29b inhibitor rescued the bacterial-killing defect. MiR-29b-mediated defects in BMT AMs were dependent on increased levels of PGE2, as miR-29b-transfected AMs treated with a novel E prostanoid receptor 2 antagonist abrogated the impaired bacterial killing. We also demonstrate that patients that have undergone HSCT exhibit increased miR-29b; thus these studies highlight miR-29b in driving defective AM responses and identify this miRNA as a potential therapeutic target.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Macrófagos Alveolares/metabolismo , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Aloinjertos , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Dinoprostona/genética , Dinoprostona/metabolismo , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
6.
J Immunol ; 190(11): 5809-17, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23630358

RESUMEN

The effectiveness of hematopoietic stem cell transplantation as a therapy for malignant and nonmalignant conditions is complicated by pulmonary infections. Using our syngeneic bone marrow transplant (BMT) mouse model, BMT mice with a reconstituted hematopoietic system displayed increased susceptibility to Pseudomonas aeruginosa and Staphylococcus aureus. BMT alveolar macrophages (AMs) exhibited a defect in P. aeruginosa phagocytosis, whereas S. aureus uptake was surprisingly enhanced. We hypothesized that the difference in phagocytosis was due to an altered scavenger receptor (SR) profile. Interestingly, MARCO expression was decreased, whereas SR-AI/II was increased. To understand how these dysregulated SR profiles might affect macrophage function, CHO cells were transfected with SR-AI/II, and phagocytosis assays revealed that SR-AI/II was important for S. aureus uptake but not for P. aeruginosa. Conversely, AMs treated in vitro with soluble MARCO exhibited similar defects in P. aeruginosa internalization as did BMT AMs. The 3'-untranslated region of SR-AI contains a putative target region for microRNA-155 (miR-155), and miR-155 expression is decreased post-BMT. Anti-miR-155-transfected AMs exhibited an increase in SR-AI/II expression and S. aureus phagocytosis. Elevated PGE2 has been implicated in driving an impaired innate immune response post-BMT. In vitro treatment of AMs with PGE2 increased SR-AI/II and decreased MARCO and miR-155. Despite a difference in phagocytic ability, BMT AMs harbor a killing defect to both P. aeruginosa and S. aureus. Thus, our data suggest that PGE2-driven alterations in SR and miR-155 expression account for the differential phagocytosis of P. aeruginosa and S. aureus, but impaired killing ultimately confers increased susceptibility to pulmonary infection.


Asunto(s)
Dinoprostona/farmacología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Fagocitosis/inmunología , Pseudomonas aeruginosa/inmunología , Receptores Depuradores/metabolismo , Staphylococcus aureus/inmunología , Animales , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Fagocitosis/genética , Infecciones por Pseudomonas/etiología , Receptores Inmunológicos/metabolismo , Receptores Depuradores/genética , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/metabolismo , Infecciones Estafilocócicas/etiología
7.
J Allergy Clin Immunol ; 133(2): 379-87, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24075232

RESUMEN

BACKGROUND: Endogenous prostanoids have been suggested to modulate sensitization during experimental allergic asthma, but the specific role of prostaglandin (PG) E2 or of specific E prostanoid (EP) receptors is not known. OBJECTIVE: Here we tested the role of EP2 signaling in allergic asthma. METHODS: Wild-type (WT) and EP2(-/-) mice were subjected to ovalbumin sensitization and acute airway challenge. The PGE2 analog misoprostol was administered during sensitization in both genotypes. In vitro culture of splenocytes and flow-sorted dendritic cells and T cells defined the mechanism by which EP2 exerted its protective effect. Adoptive transfer of WT and EP2(-/-) CD4 T cells was used to validate the importance of EP2 expression on T cells. RESULTS: Compared with WT mice, EP2(-/-) mice had exaggerated airway inflammation in this model. Splenocytes and lung lymph node cells from sensitized EP2(-/-) mice produced more IL-13 than did WT cells, suggesting increased sensitization. In WT but not EP2(-/-) mice, subcutaneous administration of misoprostol during sensitization inhibited allergic inflammation. PGE2 decreased cytokine production and inhibited signal transducer and activator of transcription 6 phosphorylation by CD3/CD28-stimulated CD4(+) T cells. Coculture of flow cytometry-sorted splenic CD4(+) T cells and CD11c(+) dendritic cells from WT or EP2(-/-) mice suggested that the increased IL-13 production in EP2(-/-) mice was due to the lack of EP2 specifically on T cells. Adoptive transfer of CD4(+) EP2(-/-) T cells caused greater cytokine production in the lungs of WT mice than did transfer of WT CD4(+) T cells. CONCLUSION: We conclude that the PGE2-EP2 axis is an important endogenous brake on allergic airway inflammation and primarily targets T cells and that its agonism represents a potential novel therapeutic approach to asthma.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Dinoprostona/inmunología , Neumonía/inmunología , Subtipo EP2 de Receptores de Prostaglandina E/inmunología , Traslado Adoptivo , Alérgenos , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Citocinas/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Noqueados , Misoprostol/farmacología , Ovalbúmina , Subtipo EP2 de Receptores de Prostaglandina E/genética , Bazo/inmunología
8.
J Immunol ; 189(9): 4528-36, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23008450

RESUMEN

Hematopoietic stem cell transplant therapy is limited by pulmonary infections. Mice with fully reconstituted hematopoietic compartments, including alveolar macrophages (AMs), after bone marrow transplantation (BMT) have impaired host defense against Gram-negative Pseudomonas aeruginosa. Impaired innate immunity is related to increased production of PGE(2) by AMs. Cyclooxygenase (COX)-2 is the rate-limiting enzyme for synthesis of PGE(2) from arachidonic acid, and COX-2 expression is elevated in AMs post-BMT. We hypothesized that epigenetic mechanisms may be responsible for upregulation of COX-2 in AMs. Using bisulfite sequencing, we observed the 5'-untranslated region and exon 1 of the COX-2 gene is hypomethylated in the AMs of BMT mice compared with control. COX-2 expression was increased in primary AMs and in the AM cell line (MHS) after treatment with 5-aza-2'-deoxycytidine (a methyltransferase inhibitor). Methylation by SssI methyltransferase of a 698-bp region of the COX-2 promoter including the beginning of exon 1 driving a luciferase reporter silenced luciferase expression. Because TGF-ß1 is elevated in lungs post-BMT, we tested whether TGF-ß1 could promote expression of COX-2 in a hypermethylated COX-2 vector, and observed TGF-ß1-induced modest expression of COX-2, suggesting an ability to demethylate the promoter. Finally, BMTs performed with marrow from mice expressing a dominant-negative form of the TGF-ßRII on CD11c-expressing cells (which includes AMs) demonstrated improved host defense and AM function. Our findings suggest impaired innate immunity and PGE(2) elevation post-BMT are due to hypomethylation of the COX-2 gene, which is at least partly regulated by TGF-ß1.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Metilación de ADN/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Regiones no Traducidas 5'/inmunología , Animales , Línea Celular , Ciclooxigenasa 2/deficiencia , Metilación de ADN/genética , Exones/inmunología , Técnicas de Silenciamiento del Gen , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología
9.
Sci Rep ; 14(1): 2033, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38263350

RESUMEN

Rapid expansion of the pulmonary microvasculature through angiogenesis drives alveolarization, the final stage of lung development that occurs postnatally and dramatically increases lung gas-exchange surface area. Disruption of pulmonary angiogenesis induces long-term structural and physiologic lung abnormalities, including bronchopulmonary dysplasia, a disease characterized by compromised alveolarization. Although endothelial cells are primary determinants of pulmonary angiogenesis, mesenchymal cells (MC) play a critical and dual role in angiogenesis and alveolarization. Therefore, we performed single cell transcriptomics and in-situ imaging of the developing lung to profile mesenchymal cells during alveolarization and in the context of lung injury. Specific mesenchymal cell subtypes were present at birth with increasing diversity during alveolarization even while expressing a distinct transcriptomic profile from more mature correlates. Hyperoxia arrested the transcriptomic progression of the MC, revealed differential cell subtype vulnerability with pericytes and myofibroblasts most affected, altered cell to cell communication, and led to the emergence of Acta1 expressing cells. These insights hold the promise of targeted treatment for neonatal lung disease, which remains a major cause of infant morbidity and mortality across the world.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Células Madre Mesenquimatosas , Recién Nacido , Lactante , Humanos , Células Endoteliales , Pulmón
10.
iScience ; 26(3): 106097, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36879800

RESUMEN

At birth, the lung is still immature, heightening susceptibility to injury but enhancing regenerative capacity. Angiogenesis drives postnatal lung development. Therefore, we profiled the transcriptional ontogeny and sensitivity to injury of pulmonary endothelial cells (EC) during early postnatal life. Although subtype speciation was evident at birth, immature lung EC exhibited transcriptomes distinct from mature counterparts, which progressed dynamically over time. Gradual, temporal changes in aerocyte capillary EC (CAP2) contrasted with more marked alterations in general capillary EC (CAP1) phenotype, including distinct CAP1 present only in the early alveolar lung expressing Peg3, a paternally imprinted transcription factor. Hyperoxia, an injury that impairs angiogenesis induced both common and unique endothelial gene signatures, dysregulated capillary EC crosstalk, and suppressed CAP1 proliferation while stimulating venous EC proliferation. These data highlight the diversity, transcriptomic evolution, and pleiotropic responses to injury of immature lung EC, possessing broad implications for lung development and injury across the lifespan.

11.
eNeuro ; 10(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36973011

RESUMEN

Cocaine-induced changes in the expression of the glutamate-related scaffolding protein Homer2 influence this drug's psychostimulant and rewarding properties. In response to neuronal activity, Homer2 is phosphorylated on S117/S216 by calcium-calmodulin kinase IIα (CaMKIIα), which induces a rapid dissociation of mGlu5-Homer2 scaffolds. Herein, we examined the requirement for Homer2 phosphorylation in cocaine-induced changes in mGlu5-Homer2 coupling, to include behavioral sensitivity to cocaine. For this, mice with alanine point mutations at (S117/216)-Homer2 (Homer2AA/AA ) were generated, and we determined their affective, cognitive and sensorimotor phenotypes, as well as cocaine-induced changes in conditioned reward and motor hyperactivity. The Homer2AA/AA mutation prevented activity-dependent phosphorylation of S216 Homer2 in cortical neurons, but Homer2AA/AA mice did not differ from wild-type (WT) controls with respect to Morris maze performance, acoustic startle, spontaneous or cocaine-induced locomotion. Homer2AA/AA mice exhibited signs of hypoanxiety similar to the phenotype of transgenic mice with a deficit in signal-regulated mGluR5 phosphorylation (Grm5AA/AA ). However, opposite of Grm5AA/AA mice, Homer2AA/AA mice were less sensitive to the aversive properties of high-dose cocaine under both place-conditioning and taste-conditioning procedures. Acute injection with cocaine caused dissociation of mGluR5 and Homer2 in striatal lysates from WT, but not Homer2AA/AA mice, suggesting a molecular basis for the deficit in cocaine aversion. These findings indicate that CaMKIIα-dependent phosphorylation of Homer2 gates the negative motivational valence of high-dose cocaine via regulation of mGlu5 binding, furthering an important role for dynamic changes in mGlu5-Homer interactions in addiction vulnerability.


Asunto(s)
Cocaína , Ratones , Animales , Cocaína/farmacología , Ratones Noqueados , Fosforilación , Ratones Transgénicos , Condicionamiento Psicológico
12.
Elife ; 92020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32484158

RESUMEN

At birth, the lungs rapidly transition from a pathogen-free, hypoxic environment to a pathogen-rich, rhythmically distended air-liquid interface. Although many studies have focused on the adult lung, the perinatal lung remains unexplored. Here, we present an atlas of the murine lung immune compartment during early postnatal development. We show that the late embryonic lung is dominated by specialized proliferative macrophages with a surprising physical interaction with the developing vasculature. These macrophages disappear after birth and are replaced by a dynamic mixture of macrophage subtypes, dendritic cells, granulocytes, and lymphocytes. Detailed characterization of macrophage diversity revealed an orchestration of distinct subpopulations across postnatal development to fill context-specific functions in tissue remodeling, angiogenesis, and immunity. These data both broaden the putative roles for immune cells in the developing lung and provide a framework for understanding how external insults alter immune cell phenotype during a period of rapid lung growth and heightened vulnerability.


Asunto(s)
Pulmón/crecimiento & desarrollo , Pulmón/inmunología , Animales , Células Dendríticas/inmunología , Granulocitos/inmunología , Homeostasis , Inmunomodulación , Linfocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Organogénesis , Fenotipo
13.
J Clin Invest ; 130(6): 3098-3112, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32134742

RESUMEN

Neutrophil accumulation is associated with lung pathology during active tuberculosis (ATB). However, the molecular mechanism or mechanisms by which neutrophils accumulate in the lung and contribute to TB immunopathology are not fully delineated. Using the well-established mouse model of TB, our new data provide evidence that the alarmin S100A8/A9 mediates neutrophil accumulation during progression to chronic TB. Depletion of neutrophils or S100A8/A9 deficiency resulted in improved Mycobacterium tuberculosis (Mtb) control during chronic but not acute TB. Mechanistically, we demonstrate that, following Mtb infection, S100A8/A9 expression is required for upregulation of the integrin molecule CD11b specifically on neutrophils, mediating their accumulation during chronic TB disease. These findings are further substantiated by increased expression of S100A8 and S100A9 mRNA in whole blood in human TB progressors when compared with nonprogressors and rapidly decreased S100A8/A9 protein levels in the serum upon TB treatment. Furthermore, we demonstrate that S100A8/A9 serum levels along with chemokines are useful in distinguishing between ATB and asymptomatic Mtb-infected latent individuals. Thus, our results support targeting S100A8/A9 pathways as host-directed therapy for TB.


Asunto(s)
Antígeno CD11b/inmunología , Calgranulina A/inmunología , Calgranulina B/inmunología , Mycobacterium tuberculosis/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Tuberculosis/inmunología , Animales , Antígeno CD11b/genética , Calgranulina A/genética , Calgranulina B/genética , Ratones , Ratones Noqueados , Neutrófilos/patología , Tuberculosis/genética , Tuberculosis/patología , Tuberculosis/terapia
14.
J Leukoc Biol ; 101(6): 1373-1381, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28258153

RESUMEN

Pulmonary tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) is a leading cause of global morbidity and mortality. The only licensed TB vaccine, Mycobacterium bovis bacillus Calmette-Guerin (BCG), has variable efficacy in protecting against pulmonary TB. Thus, the development of more effective TB vaccines is critical to control the TB epidemic. Specifically, vaccines delivered through the mucosal route are known to induce Th17 responses and provide superior protection against Mtb infection. However, already tested Th17-inducing mucosal adjuvants, such as heat-labile enterotoxins and cholera toxins, are not considered safe for use in humans. In the current study, we rationally screened adjuvants for their ability to induce Th17-polarizing cytokines in dendritic cells (DCs) and determined whether they could be used in a protective mucosal TB vaccine. Our new studies show that monophosphoryl lipid A (MPL), when used in combination with chitosan, potently induces Th17-polarizing cytokines in DCs and downstream Th17/Th1 mucosal responses and confers significant protection in mice challenged with a clinical Mtb strain. Additionally, we show that both TLRs and the inflammasome pathways are activated in DCs by MPL-chitosan to mediate induction of Th17-polarizing cytokines. Together, our studies put forward the potential of a new, protective mucosal TB vaccine candidate, which incorporates safe adjuvants already approved for use in humans.


Asunto(s)
Vacuna BCG/uso terapéutico , Membrana Mucosa/inmunología , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/prevención & control , Administración Intranasal , Animales , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/metabolismo , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología
15.
JCI Insight ; 2(19)2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28978810

RESUMEN

Mycobacterium tuberculosis (Mtb) is a global health threat, compounded by the emergence of drug-resistant strains. A hallmark of pulmonary tuberculosis (TB) is the formation of hypoxic necrotic granulomas, which upon disintegration, release infectious Mtb. Furthermore, hypoxic necrotic granulomas are associated with increased disease severity and provide a niche for drug-resistant Mtb. However, the host immune responses that promote the development of hypoxic TB granulomas are not well described. Using a necrotic Mtb mouse model, we show that loss of Mtb virulence factors, such as phenolic glycolipids, decreases the production of the proinflammatory cytokine IL-17 (also referred to as IL-17A). IL-17 production negatively regulates the development of hypoxic TB granulomas by limiting the expression of the transcription factor hypoxia-inducible factor 1α (HIF1α). In human TB patients, HIF1α mRNA expression is increased. Through genotyping and association analyses in human samples, we identified a link between the single nucleotide polymorphism rs2275913 in the IL-17 promoter (-197G/G), which is associated with decreased IL-17 production upon stimulation with Mtb cell wall. Together, our data highlight a potentially novel role for IL-17 in limiting the development of hypoxic necrotic granulomas and reducing disease severity in TB.


Asunto(s)
Granuloma/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Interleucina-17/inmunología , Tuberculosis Pulmonar/inmunología , Adulto , Anciano , Animales , Hipoxia de la Célula/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Granuloma/microbiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mediadores de Inflamación/metabolismo , Interleucina-17/biosíntesis , Masculino , Ratones Endogámicos , Persona de Mediana Edad , ARN Mensajero/genética , Tuberculosis Pulmonar/complicaciones , Adulto Joven
16.
Microbiol Spectr ; 4(5)2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27763255

RESUMEN

Chemokines and cytokines are critical for initiating and coordinating the organized and sequential recruitment and activation of cells into Mycobacterium tuberculosis-infected lungs. Correct mononuclear cellular recruitment and localization are essential to ensure control of bacterial growth without the development of diffuse and damaging granulocytic inflammation. An important block to our understanding of TB pathogenesis lies in dissecting the critical aspects of the cytokine/chemokine interplay in light of the conditional role these molecules play throughout infection and disease development. Much of the data highlighted in this review appears at first glance to be contradictory, but it is the balance between the cytokines and chemokines that is critical, and the "goldilocks" (not too much and not too little) phenomenon is paramount in any discussion of the role of these molecules in TB. Determination of how the key chemokines/cytokines and their receptors are balanced and how the loss of that balance can promote disease is vital to understanding TB pathogenesis and to identifying novel therapies for effective eradication of this disease.


Asunto(s)
Quimiocinas/inmunología , Citocinas/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Humanos , Tuberculosis/microbiología
17.
Adv Neuroimmune Biol ; 5(3): 189-197, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26709355

RESUMEN

Epigenetic regulation of gene expression is important for normal biological processes like immune cell development, immune responses, and differentiation from hematopoietic stem cells. Furthermore, it is well understood that epigenetic mechanisms can include methylation, histone modification, and more recently, microRNAs. Interestingly, aberrant epigenetic modification can also promote pathology in many diseases like cancer. The effects of methylation on gene expression and its resulting phenotype have been extensively studied. In this review, we discuss the inhibition of innate immunity that occurs in humans and animal models post-stem cell transplant. In addition, we highlight the changes methylation and microRNA profiles have on regulating pulmonary innate immune responses in the context of hematopoietic stem cell transplantation in experimental animal models.

18.
Front Immunol ; 4: 126, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23745124

RESUMEN

Infectious pulmonary complications limit the success of hematopoietic stem cell transplantation (HSCT) as a therapy for malignant and non-malignant disorders. Susceptibility to pathogens in both autologous and allogeneic HSCT recipients persists despite successful immune reconstitution. As studying the causal effects of these immune defects in the human population can be limiting, a bone marrow transplant (BMT) mouse model can be used to understand the defect in mounting a productive innate immune response post-transplantation. When syngeneic BMT is performed, this system allows the study of BMT-induced alterations in innate immune cell function that are independent of the confounding effects of immunosuppressive therapy and graft-versus-host disease. Studies from several laboratories, including our own show that pulmonary susceptibility to bacterial infections post-BMT are largely due to alterations in the lung alveolar macrophages. Changes in these cells post-BMT include cytokine and eicosanoid dysregulations, scavenger receptor alterations, changes in micro RNA profiles, and alterations in intracellular signaling molecules that limit bacterial phagocytosis and killing. The changes that occur highlight mechanisms that promote susceptibility to infections commonly afflicting HSCT recipients and provide insight into therapeutic targets that may improve patient outcomes post-HSCT.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA