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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 153(2): 362-75, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582326

RESUMEN

The functions of Nr4a1-dependent Ly6C(low) monocytes remain enigmatic. We show that they are enriched within capillaries and scavenge microparticles from their lumenal side in a steady state. In the kidney cortex, perturbation of homeostasis by a TLR7-dependent nucleic acid "danger" signal, which may signify viral infection or local cell death, triggers Gαi-dependent intravascular retention of Ly6C(low) monocytes by the endothelium. Then, monocytes recruit neutrophils in a TLR7-dependent manner to mediate focal necrosis of endothelial cells, whereas the monocytes remove cellular debris. Prevention of Ly6C(low) monocyte development, crawling, or retention in Nr4a1(-/-), Itgal(-/-), and Tlr7(host-/-BM+/+) and Cx3cr1(-/-) mice, respectively, abolished neutrophil recruitment and endothelial killing. Prevention of neutrophil recruitment in Tlr7(host+/+BM-/-) mice or by neutrophil depletion also abolished endothelial cell necrosis. Therefore, Ly6C(low) monocytes are intravascular housekeepers that orchestrate the necrosis by neutrophils of endothelial cells that signal a local threat sensed via TLR7 followed by the in situ phagocytosis of cellular debris.


Asunto(s)
Células Endoteliales/metabolismo , Monitorización Inmunológica , Monocitos/inmunología , Animales , Moléculas de Adhesión Celular/metabolismo , Micropartículas Derivadas de Células , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Humanos , Inflamación , Molécula 1 de Adhesión Intercelular/metabolismo , Riñón/irrigación sanguínea , Riñón/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Monocitos/metabolismo , Neutrófilos/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores de Quimiocina/metabolismo
2.
Nat Immunol ; 16(12): 1228-34, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26523867

RESUMEN

The molecular mechanisms that link the sympathetic stress response and inflammation remain obscure. Here we found that the transcription factor Nr4a1 regulated the production of norepinephrine (NE) in macrophages and thereby limited experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Lack of Nr4a1 in myeloid cells led to enhanced NE production, accelerated infiltration of leukocytes into the central nervous system (CNS) and disease exacerbation in vivo. In contrast, myeloid-specific deletion of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, protected mice against EAE. Furthermore, we found that Nr4a1 repressed autocrine NE production in macrophages by recruiting the corepressor CoREST to the Th promoter. Our data reveal a new role for macrophages in neuroinflammation and identify Nr4a1 as a key regulator of catecholamine production by macrophages.


Asunto(s)
Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Sistema Nervioso Simpático/inmunología , Animales , Línea Celular , Células Cultivadas , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Norepinefrina/inmunología , Norepinefrina/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sistema Nervioso Simpático/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/inmunología , Tirosina 3-Monooxigenasa/metabolismo
3.
Immunity ; 45(5): 975-987, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27814941

RESUMEN

Mononuclear phagocytes are a heterogeneous family that occupy all tissues and assume numerous roles to support tissue function and systemic homeostasis. Our ability to dissect the roles of individual subsets is limited by a lack of technologies that ablate gene function within specific mononuclear phagocyte sub-populations. Using Nr4a1-dependent Ly6Clow monocytes, we present a proof-of-principle approach that addresses these limitations. Combining ChIP-seq and molecular approaches we identified a single, conserved, sub-domain within the Nr4a1 enhancer that was essential for Ly6Clow monocyte development. Mice lacking this enhancer lacked Ly6Clow monocytes but retained Nr4a1 gene expression in macrophages during steady state and in response to LPS. Because Nr4a1 regulates inflammatory gene expression and differentiation of Ly6Clow monocytes, decoupling these processes allows Ly6Clow monocytes to be studied independently.


Asunto(s)
Diferenciación Celular/inmunología , Macrófagos/inmunología , Melanoma Experimental/inmunología , Monocitos/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Animales , Antígenos Ly/inmunología , Separación Celular , Inmunoprecipitación de Cromatina , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Reacción en Cadena de la Polimerasa
4.
Nat Immunol ; 12(8): 778-85, 2011 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-21725321

RESUMEN

The transcription factors that regulate differentiation into the monocyte subset in bone marrow have not yet been identified. Here we found that the orphan nuclear receptor NR4A1 controlled the differentiation of Ly6C- monocytes. Ly6C- monocytes, which function in a surveillance role in circulation, were absent from Nr4a1-/- mice. Normal numbers of myeloid progenitor cells were present in Nr4a1-/- mice, which indicated that the defect occurred during later stages of monocyte development. The defect was cell intrinsic, as wild-type mice that received bone marrow from Nr4a1-/- mice developed fewer patrolling monocytes than did recipients of wild-type bone marrow. The Ly6C- monocytes remaining in the bone marrow of Nr4a1-/- mice were arrested in S phase of the cell cycle and underwent apoptosis. Thus, NR4A1 functions as a master regulator of the differentiation and survival of 'patrolling' Ly6C- monocytes.


Asunto(s)
Antígenos Ly/inmunología , Apoptosis/inmunología , Médula Ósea/inmunología , Diferenciación Celular/inmunología , Monocitos/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Animales , Ciclo Celular/inmunología , Daño del ADN/inmunología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , ARN Mensajero/química , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Organismos Libres de Patógenos Específicos
5.
Mol Pharm ; 17(2): 507-516, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31841002

RESUMEN

Targeted strategies to deliver and retain drugs to kidneys are needed to improve drug accumulation and efficacy in a myriad of kidney diseases. These drug delivery systems show potential for improving the therapeutic windows of drugs acting in the kidney. Biodistribution of antibody-based therapeutics in vivo is governed by several factors including binding affinity, size, and valency. Investigations of how the biophysical and biochemical properties of biologics enable them to overcome biological barriers and reach kidneys are therefore of interest. Although renal accumulation of antibody fragments in cancer diagnostics and treatment has been observed, reports on effective delivery of antibody fragments to the kidneys remain scarce. Previously, we demonstrated that targeting plasmalemma vesicle-associated protein (PV1), a caveolae-associated protein, can promote accumulation of antibodies in both the lungs and the kidneys. Here, by fine-tuning the binding affinity of an antibody toward PV1, we observe that the anti-PV1 antibody with reduced binding affinity lost the capability for kidney targeting while retaining the lung targeting activity, suggesting that binding affinity is a critical factor for kidney targeting of the anti-PV1 antibody. We next use the antibody fragment F(ab')2 targeting PV1 to assess the dual effects of rapid kidney filtration and PV1 targeting on kidney-selective targeting. Ex vivo fluorescence imaging results demonstrated that after rapidly accumulating in kidneys at 4 h, PV1-targeted F(ab')2 was continually retained in the kidney at 24 h, whereas the isotype control F(ab')2 underwent urinary elimination with significantly reduced signaling in the kidney. Confocal imaging studies confirmed the localization of PV1-targeted F(ab')2 in the kidney. In addition, the monovalent antibody fragment (Fab-C4) lost the capability for kidney homing, indicating that the binding avidity of anti-PV1 F(ab')2 is important for kidney targeting. Our findings suggest that PV1-targeted F(ab')2 might be useful as a drug carrier for renal targeting and highlight the importance of affinity optimization for tissue targeting antibodies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Caveolas/metabolismo , Portadores de Fármacos/farmacocinética , Fragmentos Fab de Inmunoglobulinas/inmunología , Riñón/efectos de los fármacos , Proteínas de la Membrana/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Afinidad de Anticuerpos , Portadores de Fármacos/administración & dosificación , Femenino , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Riñón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
6.
Ann Rheum Dis ; 78(7): 957-966, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31040119

RESUMEN

OBJECTIVES: The presence of proinflammatory low-density granulocytes (LDG) has been demonstrated in autoimmune and infectious diseases. Recently, regulatory neutrophilic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) were identified in systemic lupus erythematosus (SLE). Because LDG and PMN-MDSC share a similar phenotype with contrasting functional effects, we explored these cells in a cohort of patients with SLE. METHODS: LDG and normal-density granulocytes (NDG) were isolated from fresh blood of healthy donors (HD) and patients with SLE. Associations between LDG and clinical manifestations were analysed. Multicolor flow cytometry and confocal imaging were performed to immunophenotype the cells. The ability of LDG and NDG to suppress T cell function and induce cytokine production was quantified. RESULTS: LDG prevalence was elevated in SLE versus HD, associated with the interferon (IFN) 21-gene signature and disease activity. Also, the LDG-to-lymphocyte ratio associated better with SLE disease activity index than neutrophil-to-lymphocyte ratio. SLE LDG exhibited significantly heightened surface expression of various activation markers and also of lectin-like oxidised low-density lipoprotein receptor-1, previously described to be associated with PMN-MDSC. Supernatants from SLE LDG did not restrict HD CD4+ T cell proliferation in an arginase-dependent manner, suggesting LDG are not immunosuppressive. SLE LDG supernatants induced proinflammatory cytokine production (IFN gamma, tumour necrosis factor alpha and lymphotoxin alpha) from CD4+ T cells. CONCLUSIONS: Based on our results, SLE LDG display an activated phenotype, exert proinflammatory effects on T cells and do not exhibit MDSC function. These results support the concept that LDG represent a distinct proinflammatory subset in SLE with pathogenic potential, at least in part, through their ability to activate type 1 helper responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Granulocitos/inmunología , Lupus Eritematoso Sistémico/inmunología , Neutrófilos/inmunología , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Proliferación Celular , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Lupus Eritematoso Sistémico/sangre , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Fenotipo , Adulto Joven
7.
Arterioscler Thromb Vasc Biol ; 35(6): 1306-16, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25838429

RESUMEN

Nonclassical patrolling monocytes are characterized by their unique ability to actively patrol the vascular endothelium under homeostatic and inflammatory conditions. Patrolling monocyte subsets (CX3CR1(high)Ly6C(-) in mouse and CX3CR1(high)CD14(dim)CD16(+) in humans) are distinct from the classical monocyte subsets (CCR2(high)Ly6C(+) in mouse and CCR2(high)CD14(+)CD16(-) in humans) and exhibit unique functions in the vasculature and inflammatory disease. Patrolling monocytes function in several disease settings to remove damaged cells and debris from the vasculature and have been associated with wound healing and the resolution of inflammation in damaged tissues. This review highlights the unique functions of these patrolling monocytes in the vasculature and during inflammation.


Asunto(s)
Endotelio Vascular/fisiopatología , Inflamación/fisiopatología , Monocitos/fisiología , Animales , Artritis/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Adhesión Celular , Diferenciación Celular , Supervivencia Celular , Humanos , Enfermedades Renales/fisiopatología , Lupus Eritematoso Sistémico/fisiopatología , Ratones , Monocitos/citología , Enfermedades del Sistema Nervioso/fisiopatología
8.
Circulation ; 127(6): 710-9, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23307829

RESUMEN

BACKGROUND: Stem cells are thought to enhance vascular remodeling in ischemic tissue in part through paracrine effects. Using molecular imaging, we tested the hypothesis that treatment of limb ischemia with multipotential adult progenitor cells (MAPCs) promotes recovery of blood flow through the recruitment of proangiogenic monocytes. METHODS AND RESULTS: Hind-limb ischemia was produced in mice by iliac artery ligation, and MAPCs were administered intramuscularly on day 1. Optical imaging of luciferase-transfected MAPCs indicated that cells survived for 1 week. Contrast-enhanced ultrasound on days 3, 7, and 21 showed a more complete recovery of blood flow and greater expansion of microvascular blood volume in MAPC-treated mice than in controls. Fluorescent microangiography demonstrated more complete distribution of flow to microvascular units in MAPC-treated mice. On ultrasound molecular imaging, expression of endothelial P-selectin and intravascular recruitment of CX(3)CR-1-positive monocytes were significantly higher in MAPC-treated mice than in the control groups at days 3 and 7 after arterial ligation. Muscle immunohistology showed a >10-fold-greater infiltration of monocytes in MAPC-treated than control-treated ischemic limbs at all time points. Intravital microscopy of ischemic or tumor necrosis factor-α-treated cremaster muscle demonstrated that MAPCs migrate to perimicrovascular locations and potentiate selectin-dependent leukocyte rolling. In vitro migration of human CD14(+) monocytes was 10-fold greater in response to MAPC-conditioned than basal media. CONCLUSIONS: In limb ischemia, MAPCs stimulate the recruitment of proangiogenic monocytes through endothelial activation and enhanced chemotaxis. These responses are sustained beyond the MAPC lifespan, suggesting that paracrine effects promote flow recovery by rebalancing the immune response toward a more regenerative phenotype.


Asunto(s)
Extremidades/irrigación sanguínea , Isquemia/terapia , Imagen Molecular , Neovascularización Fisiológica/fisiología , Comunicación Paracrina/fisiología , Trasplante de Células Madre , Células Madre Adultas/diagnóstico por imagen , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/trasplante , Animales , Receptor 1 de Quimiocinas CX3C , Movimiento Celular/fisiología , Extremidades/diagnóstico por imagen , Extremidades/patología , Humanos , Arteria Ilíaca/diagnóstico por imagen , Arteria Ilíaca/efectos de los fármacos , Arteria Ilíaca/fisiopatología , Isquemia/diagnóstico por imagen , Isquemia/patología , Receptores de Lipopolisacáridos/análisis , Ratones , Ratones Endogámicos C57BL , Microvasos/diagnóstico por imagen , Microvasos/efectos de los fármacos , Microvasos/patología , Microvasos/fisiopatología , Monocitos/patología , Monocitos/fisiología , Células Madre Multipotentes/diagnóstico por imagen , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/trasplante , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Neovascularización Fisiológica/efectos de los fármacos , Selectina-P/biosíntesis , Comunicación Paracrina/efectos de los fármacos , Receptores de Quimiocina/análisis , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/farmacología , Ultrasonografía
9.
Circ Res ; 110(3): 416-27, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22194622

RESUMEN

RATIONALE: NR4A1 (Nur77) is a nuclear receptor that is expressed in macrophages and within atherosclerotic lesions, yet its function in atherosclerosis is unknown. OBJECTIVE: Nur77 regulates the development of monocytes, particularly patrolling Ly6C(-) monocytes that may be involved in resolution of inflammation. We sought to determine how absence of nuclear receptor subfamily 4, group A, member 1 (NR4A1) in hematopoietic cells affected atherosclerosis development. METHODS AND RESULTS: Nur77(-/-) chimeric mice on a Ldlr(-/-) background showed a 3-fold increase in atherosclerosis development when fed a Western diet for 20 weeks, despite having a drastic reduction in Ly6C(-) patrolling monocytes. In a second model, mice deficient in both Nur77 and ApoE (ApoE(-/-)Nur77(-/-)) also showed increased atherosclerosis after 11 weeks of Western diet. Atherosclerosis was associated with a significant change in macrophage polarization toward a proinflammatory phenotype, with high expression of tumor necrosis factor-α and nitric oxide and low expression of Arginase-I. Moreover, we found increased expression of toll-like receptor 4 mRNA and protein in Nur77(-/-) macrophages as well as increased phosphorylation of the p65 subunit of NFκB. Inhibition of NFκB activity blocked excess activation of Nur77(-/-) macrophages. CONCLUSIONS: We conclude that the absence of Nur77 in monocytes and macrophages results in enhanced toll-like receptor signaling and polarization of macrophages toward a proinflammatory M1 phenotype. Despite having fewer monocytes, Nur77(-/-) mice developed significant atherosclerosis when fed a Western diet. These studies indicate that Nur77 is a novel target for modulating the inflammatory phenotype of monocytes and macrophages and may be important for regulation of atherogenesis.


Asunto(s)
Aterosclerosis/patología , Eliminación de Gen , Inflamación/patología , Macrófagos/patología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Fenotipo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Aterosclerosis/etiología , Aterosclerosis/fisiopatología , Dieta/efectos adversos , Modelos Animales de Enfermedad , Humanos , Inflamación/fisiopatología , Metabolismo de los Lípidos/fisiología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores de LDL/fisiología , Receptores Toll-Like/fisiología
10.
Curr Opin Lipidol ; 24(5): 381-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24005216

RESUMEN

PURPOSE OF REVIEW: To understand chronic inflammatory diseases such as atherosclerosis, we require in-depth knowledge on immune-cell differentiation, function of specific immune-cell subsets and endothelial cell-mediated extravasation. In this review, we summarize a number of very recent observations on the pivotal function of NR4A nuclear receptors in immunity and atherosclerosis. RECENT FINDINGS: NR4A nuclear receptors are involved in negative selection of thymocytes, Treg differentiation and the development of Ly6C monocytes. Nur77 and Nurr1 attenuate atherosclerosis in mice whereas NOR-1 aggravates vascular lesion formation. SUMMARY: These exciting, novel insights on the function of NR4A nuclear receptors in immunity, vascular cells and atherosclerosis will initiate a plethora of studies to understand the underlying molecular mechanisms, which will culminate in the identification of novel NR4A targets to modulate chronic inflammatory disease.


Asunto(s)
Aterosclerosis/inmunología , Células Endoteliales/inmunología , Monocitos/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Linfocitos T Reguladores/inmunología , Animales , Aterosclerosis/patología , Diferenciación Celular/inmunología , Células Endoteliales/patología , Humanos , Ratones , Monocitos/patología , Linfocitos T Reguladores/patología
11.
Front Genet ; 13: 868015, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711912

RESUMEN

Target prioritization is essential for drug discovery and repositioning. Applying computational methods to analyze and process multi-omics data to find new drug targets is a practical approach for achieving this. Despite an increasing number of methods for generating datasets such as genomics, phenomics, and proteomics, attempts to integrate and mine such datasets remain limited in scope. Developing hybrid intelligence solutions that combine human intelligence in the scientific domain and disease biology with the ability to mine multiple databases simultaneously may help augment drug target discovery and identify novel drug-indication associations. We believe that integrating different data sources using a singular numerical scoring system in a hybrid intelligent framework could help to bridge these different omics layers and facilitate rapid drug target prioritization for studies in drug discovery, development or repositioning. Herein, we describe our prototype of the StarGazer pipeline which combines multi-source, multi-omics data with a novel target prioritization scoring system in an interactive Python-based Streamlit dashboard. StarGazer displays target prioritization scores for genes associated with 1844 phenotypic traits, and is available via https://github.com/AstraZeneca/StarGazer.

12.
Biol Reprod ; 82(1): 112-22, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19741205

RESUMEN

The zebrafish nuclear progestin receptor (nPR; official symbol PGR) was identified and characterized to better understand its role in regulating reproduction in this well-established teleost model. A full-length cDNA was identified that encoded a 617-amino acid residue protein with high homology to PGRs in other vertebrates, and contained five domains characteristic of nuclear steroid receptors. In contrast to the multiplicity of steroid receptors often found in euteleosts and attributed to probable genome duplication, only a single locus encoding the full-length zebrafish pgr was identified. Cytosolic proteins from pgr-transfected cells showed a high affinity (K(d) = 2 nM), saturable, single-binding site specific for a native progestin in euteleosts, 4-pregnen-17,20 beta-diol-3-one (17,20 beta-DHP). Both 17,20 beta-DHP and progesterone were potent inducers of transcriptional activity in cells transiently transfected with pgr in a dual luciferase reporter assay, whereas androgens and estrogens had little potency. The pgr transcript and protein were abundant in the ovaries, testis, and brain and were scarce or undetectable in the intestine, muscle, and gills. Further analyses indicate that Pgr was expressed robustly in the preoptic region of the hypothalamus in the brain; proliferating spermatogonia and early spermatocytes in the testis; and in follicular cells and early-stage oocytes (stages I and II), with very low levels within maturationally competent late-stage oocytes (IV) in the ovary. The localization of Pgr suggests that it mediates progestin regulation of reproductive signaling in the brain, early germ cell proliferation in testis, and ovarian follicular functions, but not final oocyte or sperm maturation.


Asunto(s)
Oocitos/metabolismo , Área Preóptica/metabolismo , Receptores de Progesterona/metabolismo , Testículo/metabolismo , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Proliferación Celular , Cartilla de ADN , Femenino , Hidroxiprogesteronas/metabolismo , Inmunohistoquímica , Masculino , Datos de Secuencia Molecular , Receptores de Progesterona/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
13.
J Leukoc Biol ; 107(6): 883-892, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32386455

RESUMEN

Nonclassical monocytes maintain vascular homeostasis by patrolling the vascular endothelium, responding to inflammatory signals, and scavenging cellular debris. Nonclassical monocytes also prevent metastatic tumor cells from seeding new tissues, but whether the patrolling function of nonclassical monocytes is required for this process is unknown. To answer this question, we utilized an inducible-knockout mouse that exhibits loss of the integrin-adaptor protein Kindlin-3 specifically in nonclassical monocytes. We show that Kindlin-3-deficient nonclassical monocytes are unable to patrol the vascular endothelium in either the lungs or periphery. We also find that Kindlin-3-deficient nonclassical monocytes cannot firmly adhere to, and instead "slip" along, the vascular endothelium. Loss of patrolling activity by nonclassical monocytes was phenocopied by ablation of LFA-1, an integrin-binding partner of Kindlin-3. When B16F10 murine melanoma tumor cells were introduced into Kindlin-3-deficient mice, nonclassical monocytes showed defective patrolling towards tumor cells and failure to ingest tumor particles in vivo. Consequently, we observed a significant, 4-fold increase in lung tumor metastases in mice possessing Kindlin-3-deficient nonclassical monocytes. Thus, we conclude that the patrolling function of nonclassical monocytes is mediated by Kindlin-3 and essential for these cells to maintain vascular endothelial homeostasis and prevent tumor metastasis to the lung.


Asunto(s)
Proteínas del Citoesqueleto/genética , Regulación Neoplásica de la Expresión Génica , Antígeno-1 Asociado a Función de Linfocito/genética , Melanoma Experimental/genética , Monocitos/inmunología , Fagocitosis , Neoplasias Cutáneas/genética , Animales , Médula Ósea/inmunología , Trasplante de Médula Ósea , Adhesión Celular , Comunicación Celular/inmunología , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Femenino , Humanos , Inyecciones Intravenosas , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/secundario , Ratones , Ratones Noqueados , Monocitos/patología , Células Neoplásicas Circulantes/inmunología , Células Neoplásicas Circulantes/patología , Cultivo Primario de Células , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Irradiación Corporal Total
14.
ACS Chem Biol ; 15(4): 830-836, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32155049

RESUMEN

Efficacious use of therapeutic gene delivery via nanoparticles is hampered by the challenges associated with targeted delivery to tissues of interest. Systemic administration of lipid nanoparticle (LNP)-encapsulated mRNA leads to a protein expressed predominantly in the liver and spleen. Here, LNP encapsulating mRNA was covalently conjugated to an antibody, specifically binding plasmalemma vesicle-associated protein (PV1) as a means to target lung tissue. Systemic administration of PV1-targeted LNPs demonstrated significantly increased delivery of mRNA to the lungs and a 40-fold improvement in protein expression in the lungs, compared with control LNPs. We also investigated the effect of LNP size to determine optimal tissue distribution and transfection. Larger-size PV1-targeted LNPs not only have the elasticity to target the PV1 expressed in the caveolae but also enable robust mRNA expression in the lungs. Targeted delivery of mRNA to the lungs is a promising approach in the treatment of lung diseases.


Asunto(s)
Portadores de Fármacos/química , Lípidos/química , Pulmón/metabolismo , Nanopartículas/química , ARN Mensajero/farmacología , Animales , Anticuerpos Inmovilizados/química , Anticuerpos Inmovilizados/inmunología , Caveolas/inmunología , Femenino , Técnicas de Transferencia de Gen , Proteínas de la Membrana/inmunología , Ratones Endogámicos BALB C
15.
JCI Insight ; 5(17)2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32721945

RESUMEN

Dysregulated sensing of self-nucleic acid is a leading cause of autoimmunity in multifactorial and monogenic diseases. Mutations in Wiskott-Aldrich syndrome protein (WASp), a key regulator of cytoskeletal dynamics in immune cells, cause autoimmune manifestations and increased production of type I IFNs by innate cells. Here we show that immune complexes of self-DNA and autoantibodies (DNA-ICs) contribute to elevated IFN levels via activation of the cGAS/STING pathway of cytosolic sensing. Mechanistically, lack of endosomal F-actin nucleation by WASp caused a delay in endolysosomal maturation and prolonged the transit time of ingested DNA-ICs. Stalling in maturation-defective organelles facilitated leakage of DNA-ICs into the cytosol, promoting activation of the TBK1/STING pathway. Genetic deletion of STING and STING and cGAS chemical inhibitors abolished IFN production and rescued systemic activation of IFN-stimulated genes in vivo. These data unveil the contribution of cytosolic self-nucleic acid sensing in WAS and underscore the importance of WASp-mediated endosomal actin remodeling in preventing innate activation.


Asunto(s)
ADN/inmunología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Animales , Autoanticuerpos/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Endosomas/metabolismo , Inmunidad Innata , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo
16.
Gen Comp Endocrinol ; 161(1): 153-7, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18957293

RESUMEN

A distinct family of membrane progestin receptors (mPRalpha, mPRbeta, and mPRgamma) that mediate rapid, nongenomic actions of progestins has been identified and characterized in several fish species as well as in frogs, rats, pigs, and humans. However, few studies to date have thoroughly examined tissue specific expression of mPR protein and transcripts in any model species. In the present study, the expression of both mPRalpha and mPRbeta in zebrafish was examined by RT-PCR, Western blotting and immunohistochemistry using mPR specific primers and antibodies. The proteins and mRNAs of mPRalpha and mPRbeta were co-localized in the major reproductive organs, including the ovary, testis, and pituitary. Both mPRalpha and mPRbeta were found in scattered cells in the pituitary for the first time. In the testis, immunostaining of mPRalpha was restricted to the sperm, while mPRbeta was found in spermatocytes and spermatogonia. In the ovary, both mPRalpha and mPRbeta were detected in denude oocytes and follicular layer cells. Furthermore, mPRalpha and mPRbeta proteins were localized at or near the oocyte membrane of maturationally competent stage IV oocytes in a probable location for mediating progestin-induced nongenomic signaling in oocytes.


Asunto(s)
Oocitos/metabolismo , Hipófisis/metabolismo , Receptores de Progesterona/genética , Testículo/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Inmunohistoquímica , Masculino , Pez Cebra/genética
17.
Commun Biol ; 2: 92, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30854484

RESUMEN

Systemic administration of bio-therapeutics can result in only a fraction of drug reaching targeted tissues, with the majority of drug being distributed to tissues irrelevant to the drug's site of action. Targeted delivery to specific organs may allow for greater accumulation, better efficacy, and improved safety. We investigated how targeting plasmalemma vesicle-associated protein (PV1), a protein found in the endothelial caveolae of lungs and kidneys, can promote accumulation in these organs. Using ex vivo fluorescence imaging, we show that intravenously administered αPV1 antibodies localize to mouse lungs and kidneys. In a bleomycin-induced idiopathic pulmonary fibrosis (IPF) mouse model, αPV1 conjugated to Prostaglandin E2 (PGE2), a known anti-fibrotic agent, significantly reduced collagen content and fibrosis whereas a non-targeted PGE2 antibody conjugate failed to slow fibrosis progression. Our results demonstrate that PV1 targeting can be utilized to deliver therapeutics to lungs and this approach is potentially applicable for various lung diseases.


Asunto(s)
Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Proteínas de la Membrana/metabolismo , Animales , Biomarcadores , Bleomicina/efectos adversos , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones
18.
J Clin Invest ; 128(5): 1873-1887, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29611821

RESUMEN

Uncontrolled secretion of type I IFN, as the result of endosomal TLR (i.e., TLR7 and TLR9) signaling in plasmacytoid DCs (pDCs), and abnormal production of autoantibodies by B cells are critical for systemic lupus erythematosus (SLE) pathogenesis. The importance of galectin-9 (Gal-9) in regulating various autoimmune diseases, including lupus, has been demonstrated. However, the precise mechanism by which Gal-9 mediates this effect remains unclear. Here, using spontaneous murine models of lupus (i.e., BXSB/MpJ and NZB/W F1 mice), we demonstrate that administration of Gal-9 results in reduced TLR7-mediated autoimmune manifestations. While investigating the mechanism underlying this phenomenon, we observed that Gal-9 inhibits the phenotypic maturation of pDCs and B cells and abrogates their ability to mount cytokine responses to TLR7/TLR9 ligands. Importantly, immunocomplex-mediated (IC-mediated) and neutrophil extracellular trap-mediated (NET-mediated) pDC activation was inhibited by Gal-9. Additionally, the mTOR/p70S6K pathway, which is recruited by both pDCs and B cells for TLR-mediated IFN secretion and autoantibody generation, respectively, was attenuated. Gal-9 was found to exert its inhibitory effect on both the cells by interacting with CD44.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/inmunología , Células Dendríticas/inmunología , Galectinas/inmunología , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/inmunología , Receptor Toll-Like 7/inmunología , Animales , Linfocitos B/patología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Lupus Eritematoso Sistémico/patología , Masculino , Ratones , Proteínas Quinasas S6 Ribosómicas 70-kDa/inmunología , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/inmunología , Receptor Toll-Like 9/inmunología
19.
Lupus Sci Med ; 5(1): e000261, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29644082

RESUMEN

OBJECTIVE: We investigated the mechanistic and pharmacological properties of anifrolumab, a fully human, effector-null, anti-type I interferon (IFN) alpha receptor 1 (IFNAR1) monoclonal antibody in development for SLE. METHODS: IFNAR1 surface expression and internalisation on human monocytes before and after exposure to anifrolumab were assessed using confocal microscopy and flow cytometry. The effects of anifrolumab on type I IFN pathway activation were assessed using signal transducer and activator of transcription 1 (STAT1) phosphorylation, IFN-stimulated response element-luciferase reporter cell assays and type I IFN gene signature induction. The ability of anifrolumab to inhibit plasmacytoid dendritic cell (pDC) function and plasma cell differentiation was assessed by flow cytometry and ELISA. Effector-null properties of anifrolumab were assessed in antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays with B cells. RESULTS: Anifrolumab reduced cell surface IFNAR1 by eliciting IFNAR1 internalisation. Anifrolumab blocked type I IFN-dependent STAT1 phosphorylation and IFN-dependent signalling induced by recombinant and pDC-derived type I IFNs and serum of patients with SLE. Anifrolumab suppressed type I IFN production by blocking the type I IFN autoamplification loop and inhibited proinflammatory cytokine induction and the upregulation of costimulatory molecules on stimulated pDCs. Blockade of IFNAR1 suppressed plasma cell differentiation in pDC/B cell co-cultures. Anifrolumab did not exhibit CDC or ADCC activity. CONCLUSIONS: Anifrolumab potently inhibits type I IFN-dependent signalling, including the type I IFN autoamplification loop, and is a promising therapeutic for patients with SLE and other diseases that exhibit chronic dysfunctional type I IFN signalling.

20.
Bio Protoc ; 6(20)2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29552586

RESUMEN

Extracellular tumor material including exosomes, microvesicles and apoptotic tumor debris may help cancers invade new organs. Enhancing the removal of extracellular tumor material by immune cells represents a novel immunotherapy approach for preventing cancer metastasis. This protocol quantifies the uptake and removal of extracellular tumor material from circulation and tissues by immune cells. In this assay fluorescent tumor cells are transferred into mice, and then immune cells are quantified by either flow cytometry or imaging cytometry for their uptake of tumor material.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA