Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(6): 1634-1650.e17, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30433869

RESUMEN

Innate immune memory is an emerging area of research. However, innate immune memory at major mucosal sites remains poorly understood. Here, we show that respiratory viral infection induces long-lasting memory alveolar macrophages (AMs). Memory AMs are programed to express high MHC II, a defense-ready gene signature, and increased glycolytic metabolism, and produce, upon re-stimulation, neutrophil chemokines. Using a multitude of approaches, we reveal that the priming, but not maintenance, of memory AMs requires the help from effector CD8 T cells. T cells jump-start this process via IFN-γ production. We further find that formation and maintenance of memory AMs are independent of monocytes or bone marrow progenitors. Finally, we demonstrate that memory AMs are poised for robust trained immunity against bacterial infection in the lung via rapid induction of chemokines and neutrophilia. Our study thus establishes a new paradigm of immunological memory formation whereby adaptive T-lymphocytes render innate memory of mucosal-associated macrophages.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Innata , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos T CD8-positivos/citología , Memoria Inmunológica , Pulmón/citología , Macrófagos Alveolares/citología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Monocitos/citología , Monocitos/inmunología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Linfocitos T Colaboradores-Inductores/citología
2.
Cell ; 172(1-2): 176-190.e19, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29328912

RESUMEN

The dogma that adaptive immunity is the only arm of the immune response with memory capacity has been recently challenged by several studies demonstrating evidence for memory-like innate immune training. However, the underlying mechanisms and location for generating such innate memory responses in vivo remain unknown. Here, we show that access of Bacillus Calmette-Guérin (BCG) to the bone marrow (BM) changes the transcriptional landscape of hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs), leading to local cell expansion and enhanced myelopoiesis at the expense of lymphopoiesis. Importantly, BCG-educated HSCs generate epigenetically modified macrophages that provide significantly better protection against virulent M. tuberculosis infection than naïve macrophages. By using parabiotic and chimeric mice, as well as adoptive transfer approaches, we demonstrate that training of the monocyte/macrophage lineage via BCG-induced HSC reprogramming is sustainable in vivo. Our results indicate that targeting the HSC compartment provides a novel approach for vaccine development.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inmunidad Innata , Memoria Inmunológica , Mycobacterium bovis/inmunología , Transcriptoma , Animales , Línea Celular , Células Cultivadas , Epigénesis Genética , Hematopoyesis , Ratones , Ratones Endogámicos C57BL , Tuberculosis/inmunología
3.
Cell ; 172(5): 1050-1062.e14, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474906

RESUMEN

While the preponderance of morbidity and mortality in medulloblastoma patients are due to metastatic disease, most research focuses on the primary tumor due to a dearth of metastatic tissue samples and model systems. Medulloblastoma metastases are found almost exclusively on the leptomeningeal surface of the brain and spinal cord; dissemination is therefore thought to occur through shedding of primary tumor cells into the cerebrospinal fluid followed by distal re-implantation on the leptomeninges. We present evidence for medulloblastoma circulating tumor cells (CTCs) in therapy-naive patients and demonstrate in vivo, through flank xenografting and parabiosis, that medulloblastoma CTCs can spread through the blood to the leptomeningeal space to form leptomeningeal metastases. Medulloblastoma leptomeningeal metastases express high levels of the chemokine CCL2, and expression of CCL2 in medulloblastoma in vivo is sufficient to drive leptomeningeal dissemination. Hematogenous dissemination of medulloblastoma offers a new opportunity to diagnose and treat lethal disseminated medulloblastoma.


Asunto(s)
Meduloblastoma/irrigación sanguínea , Meduloblastoma/patología , Neoplasias Meníngeas/irrigación sanguínea , Neoplasias Meníngeas/secundario , Aloinjertos , Animales , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Cromosomas Humanos Par 10/genética , Femenino , Humanos , Masculino , Meduloblastoma/genética , Ratones SCID , Células Neoplásicas Circulantes , Parabiosis
4.
Nat Immunol ; 20(1): 29-39, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30538339

RESUMEN

Macrophages promote both injury and repair after myocardial infarction, but discriminating functions within mixed populations remains challenging. Here we used fate mapping, parabiosis and single-cell transcriptomics to demonstrate that at steady state, TIMD4+LYVE1+MHC-IIloCCR2- resident cardiac macrophages self-renew with negligible blood monocyte input. Monocytes partially replaced resident TIMD4-LYVE1-MHC-IIhiCCR2- macrophages and fully replaced TIMD4-LYVE1-MHC-IIhiCCR2+ macrophages, revealing a hierarchy of monocyte contribution to functionally distinct macrophage subsets. Ischemic injury reduced TIMD4+ and TIMD4- resident macrophage abundance, whereas CCR2+ monocyte-derived macrophages adopted multiple cell fates within infarcted tissue, including those nearly indistinguishable from resident macrophages. Recruited macrophages did not express TIMD4, highlighting the ability of TIMD4 to track a subset of resident macrophages in the absence of fate mapping. Despite this similarity, inducible depletion of resident macrophages using a Cx3cr1-based system led to impaired cardiac function and promoted adverse remodeling primarily within the peri-infarct zone, revealing a nonredundant, cardioprotective role of resident cardiac macrophages.


Asunto(s)
Macrófagos/fisiología , Infarto del Miocardio/inmunología , Miocardio/patología , Animales , Receptor 1 de Quimiocinas CX3C/metabolismo , Diferenciación Celular , Linaje de la Célula , Autorrenovación de las Células , Perfilación de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Parabiosis , Receptores CCR2/genética , Receptores CCR2/metabolismo , Análisis de la Célula Individual , Remodelación Ventricular , Proteínas de Transporte Vesicular/metabolismo
5.
Nat Immunol ; 20(5): 664, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30862954

RESUMEN

In the version of this article initially published, the equal contribution of the third author was omitted. The footnote links for that author should be "Sara Nejat1,11" and the correct statement is as follows: "11These authors contributed equally: Sarah A. Dick, Jillian A. Macklin, Sara Nejat." The error has been corrected in the HTML and PDF versions of the article.

6.
Immunity ; 55(5): 862-878.e8, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35508166

RESUMEN

Macrophage colony stimulating factor-1 (CSF-1) plays a critical role in maintaining myeloid lineage cells. However, congenital global deficiency of CSF-1 (Csf1op/op) causes severe musculoskeletal defects that may indirectly affect hematopoiesis. Indeed, we show here that osteolineage-derived Csf1 prevented developmental abnormalities but had no effect on monopoiesis in adulthood. However, ubiquitous deletion of Csf1 conditionally in adulthood decreased monocyte survival, differentiation, and migration, independent of its effects on bone development. Bone histology revealed that monocytes reside near sinusoidal endothelial cells (ECs) and leptin receptor (Lepr)-expressing perivascular mesenchymal stromal cells (MSCs). Targeted deletion of Csf1 from sinusoidal ECs selectively reduced Ly6C- monocytes, whereas combined depletion of Csf1 from ECs and MSCs further decreased Ly6Chi cells. Moreover, EC-derived CSF-1 facilitated recovery of Ly6C- monocytes and protected mice from weight loss following induction of polymicrobial sepsis. Thus, monocytes are supported by distinct cellular sources of CSF-1 within a perivascular BM niche.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos , Células Madre Mesenquimatosas , Animales , Médula Ósea , Células de la Médula Ósea , Células Endoteliales , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Monocitos
8.
Nat Immunol ; 17(11): 1263-1272, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27668800

RESUMEN

Regions of the normal arterial intima predisposed to atherosclerosis are sites of ongoing monocyte trafficking and also contain resident myeloid cells with features of dendritic cells. However, the pathophysiological roles of these cells are poorly understood. Here we found that intimal myeloid cells underwent reverse transendothelial migration (RTM) into the arterial circulation after systemic stimulation of pattern-recognition receptors (PRRs). This process was dependent on expression of the chemokine receptor CCR7 and its ligand CCL19 by intimal myeloid cells. In mice infected with the intracellular pathogen Chlamydia muridarum, blood monocytes disseminated infection to the intima. Subsequent CCL19-CCR7-dependent RTM was critical for the clearance of intimal C. muridarum. This process was inhibited by hypercholesterolemia. Thus, RTM protects the normal arterial intima, and compromised RTM during atherogenesis might contribute to the intracellular retention of pathogens in atherosclerotic lesions.


Asunto(s)
Quimiocina CCL19/metabolismo , Chlamydia muridarum/inmunología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Receptores CCR7/metabolismo , Migración Transendotelial y Transepitelial , Túnica Íntima/inmunología , Túnica Íntima/metabolismo , Animales , Antígeno CD11c/metabolismo , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/virología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/microbiología , ARN Mensajero/genética , Transducción de Señal , Receptores Toll-Like/metabolismo , Túnica Íntima/microbiología
9.
Nat Immunol ; 17(2): 159-68, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26642357

RESUMEN

Resident macrophages densely populate the normal arterial wall, yet their origins and the mechanisms that sustain them are poorly understood. Here we use gene-expression profiling to show that arterial macrophages constitute a distinct population among macrophages. Using multiple fate-mapping approaches, we show that arterial macrophages arise embryonically from CX3CR1(+) precursors and postnatally from bone marrow-derived monocytes that colonize the tissue immediately after birth. In adulthood, proliferation (rather than monocyte recruitment) sustains arterial macrophages in the steady state and after severe depletion following sepsis. After infection, arterial macrophages return rapidly to functional homeostasis. Finally, survival of resident arterial macrophages depends on a CX3CR1-CX3CL1 axis within the vascular niche.


Asunto(s)
Autorrenovación de las Células , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Monocitos/citología , Monocitos/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C , Supervivencia Celular , Quimiocina CX3CL1/metabolismo , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Inmunofenotipificación , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Unión Proteica , Nicho de Células Madre , Transcriptoma
11.
Immunity ; 47(5): 974-989.e8, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166591

RESUMEN

Innate and adaptive immune cells modulate heart failure pathogenesis during viral myocarditis, yet their identities and functions remain poorly defined. We utilized a combination of genetic fate mapping, parabiotic, transcriptional, and functional analyses and demonstrated that the heart contained two major conventional dendritic cell (cDC) subsets, CD103+ and CD11b+, which differentially relied on local proliferation and precursor recruitment to maintain their tissue residency. Following viral infection of the myocardium, cDCs accumulated in the heart coincident with monocyte infiltration and loss of resident reparative embryonic-derived cardiac macrophages. cDC depletion abrogated antigen-specific CD8+ T cell proliferative expansion, transforming subclinical cardiac injury to overt heart failure. These effects were mediated by CD103+ cDCs, which are dependent on the transcription factor BATF3 for their development. Collectively, our findings identified resident cardiac cDC subsets, defined their origins, and revealed an essential role for CD103+ cDCs in antigen-specific T cell responses during subclinical viral myocarditis.


Asunto(s)
Antígenos CD/análisis , Infecciones por Cardiovirus/complicaciones , Células Dendríticas/inmunología , Virus de la Encefalomiocarditis , Insuficiencia Cardíaca/prevención & control , Cadenas alfa de Integrinas/análisis , Miocarditis/complicaciones , Animales , Antígeno CD11b/análisis , Linfocitos T CD8-positivos/inmunología , Infecciones por Cardiovirus/inmunología , Movimiento Celular , Femenino , Hematopoyesis , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Miocarditis/inmunología , Receptores CCR2/fisiología
13.
Small ; 19(41): e2302917, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37312676

RESUMEN

Lipid nanoparticles (LNPs) and ribonucleic acid (RNA) technology are highly versatile tools that can be deployed for diagnostic, prophylactic, and therapeutic applications. In this report, supramolecular chemistry concepts are incorporated into the rational design of a new ionizable lipid, C3-K2-E14, for systemic administration. This lipid incorporates a cone-shaped structure intended to facilitate cell bilayer disruption, and three tertiary amines to improve RNA binding. Additionally, hydroxyl and amide motifs are incorporated to further enhance RNA binding and improve LNP stability. Optimization of messenger RNA (mRNA) and small interfering RNA (siRNA) formulation conditions and lipid ratios produce LNPs with favorable diameter (<150 nm), polydispersity index (<0.15), and RNA encapsulation efficiency (>90%), all of which are preserved after 2 months at 4 or 37 °C storage in ready-to-use liquid form. The lipid and formulated LNPs are well-tolerated in animals and show no deleterious material-induced effects. Furthermore, 1 week after intravenous LNP administration, fluorescent signal from tagged RNA payloads are not detected. To demonstrate the long-term treatment potential for chronic diseases, repeated dosing of C3-K2-E14 LNPs containing siRNA that silences the colony stimulating factor-1 (CSF-1) gene can modulate leukocyte populations in vivo, further highlighting utility.


Asunto(s)
Nanopartículas , Animales , ARN Interferente Pequeño , ARN Mensajero/genética , Nanopartículas/química , Lípidos/química
14.
Circ Res ; 128(4): 530-543, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33397122

RESUMEN

RATIONALE: Bone marrow transplantation (BMT) is used frequently to study the role of hematopoietic cells in atherosclerosis, but aortic arch lesions are smaller in mice after BMT. OBJECTIVE: To identify the earliest stage of atherosclerosis inhibited by BMT and elucidate potential mechanisms. METHODS AND RESULTS: Ldlr-/- mice underwent total body γ-irradiation, bone marrow reconstitution, and 6-week recovery. Atherosclerosis was studied in the ascending aortic arch and compared with mice without BMT. In BMT mice, neutral lipid and myeloid cell topography were lower in lesions after feeding a cholesterol-rich diet for 3, 6, and 12 weeks. Lesion coalescence and height were suppressed dramatically in mice post-BMT, whereas lateral growth was inhibited minimally. Targeted radiation to the upper thorax alone reproduced the BMT phenotype. Classical monocyte recruitment, intimal myeloid cell proliferation, and apoptosis did not account for the post-BMT phenotype. Neutral lipid accumulation was reduced in 5-day lesions, thus we developed quantitative assays for LDL (low-density lipoprotein) accumulation and paracellular leakage using DiI-labeled human LDL and rhodamine B-labeled 70 kD dextran. LDL accumulation was dramatically higher in the intima of Ldlr-/- relative to Ldlr+/+ mice, and was inhibited by injection of HDL mimics, suggesting a regulated process. LDL, but not dextran, accumulation was lower in mice post-BMT both at baseline and in 5-day lesions. Since the transcript abundance of molecules implicated in LDL transcytosis was not significantly different in the post-BMT intima, transcriptomics from whole aortic arch intima, and at single-cell resolution, was performed to give insights into pathways modulated by BMT. CONCLUSIONS: Radiation exposure inhibits LDL entry into the aortic intima at baseline and the earliest stages of atherosclerosis. Single-cell transcriptomic analysis suggests that LDL uptake by endothelial cells is diverted to lysosomal degradation and reverse cholesterol transport pathways. This reduces intimal accumulation of lipid and impacts lesion initiation and growth.


Asunto(s)
Aterosclerosis/metabolismo , Rayos gamma , Lipoproteínas LDL/metabolismo , Túnica Íntima/efectos de la radiación , Animales , Aorta/metabolismo , Aorta/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transcriptoma , Túnica Íntima/metabolismo
15.
Clin Sci (Lond) ; 135(10): 1295-1309, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33978148

RESUMEN

OBJECTIVE: Aortic macrophage accumulation is characteristic of the pathogenesis of abdominal aortic aneurysm (AAA) but the mechanisms of macrophage accumulation and their phenotype are poorly understood. Lymphatic vessel endothelial receptor-1 (Lyve-1+) resident aortic macrophages independently self-renew and are functionally distinct from monocyte-derived macrophages recruited during inflammation. We hypothesized that Lyve-1+ and Lyve-1- macrophages differentially contribute to aortic aneurysm. Approach and results: Angiotensin-2 and ß-aminopropionitrile (AT2/BAPN) were administered to induce AAA in C57BL/6J mice. Using immunohistochemistry (IHC), we demonstrated primarily adventitial accumulation of aortic macrophages, and in association with areas of elastin fragmentation and aortic dissection. Compared with controls, AAA was associated with a relative percent depletion of Lyve-1+ resident aortic macrophages and accumulation of Lyve-1- macrophages. Using CD45.1/CD45.2 parabiosis, we demonstrated aortic macrophage recruitment in AAA. Depletion of aortic macrophages in CCR2-/- mice was associated with reduced aortic dilatation indicating the functional role of recruitment from the bone marrow. Depletion of aortic macrophages using anti-macrophage colony-stimulating factor 1 receptor (MCSF1R)-neutralizing antibody (Ab) reduced the incidence of AAA. Conditional depletion of Lyve-1+ aortic macrophages was achieved by generating Lyve-1wt/cre Csf1rfl/fl mice. Selective depletion of Lyve-1+ aortic macrophages had no protective effects following AT2/BAPN administration and resulted in increased aortic dilatation in the suprarenal aorta. CONCLUSIONS: Aortic macrophage accumulation in AAA derives from adventitial recruitment of Lyve-1- macrophages, with relative percent depletion of Lyve-1+ macrophages. Selective targeting of macrophage subtypes represents a potential novel therapeutic avenue for the medical treatment of AAA.


Asunto(s)
Angiotensina II/metabolismo , Aorta Abdominal/metabolismo , Macrófagos/inmunología , Proteínas de Transporte de Membrana/metabolismo , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/patología , Aneurisma de la Aorta/patología , Aneurisma de la Aorta Abdominal/patología , Modelos Animales de Enfermedad , Inflamación/patología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Proteínas de Transporte de Membrana/inmunología , Ratones , Transducción de Señal/inmunología
16.
Basic Res Cardiol ; 115(6): 78, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33296022

RESUMEN

Statins induce plaque regression characterized by reduced macrophage content in humans, but the underlying mechanisms remain speculative. Studying the translational APOE*3-Leiden.CETP mouse model with a humanized lipoprotein metabolism, we find that systemic cholesterol lowering by oral atorvastatin or dietary restriction inhibits monocyte infiltration, and reverses macrophage accumulation in atherosclerotic plaques. Contrary to current believes, none of (1) reduced monocyte influx (studied by cell fate mapping in thorax-shielded irradiation bone marrow chimeras), (2) enhanced macrophage egress (studied by fluorescent bead labeling and transfer), or (3) atorvastatin accumulation in murine or human plaque (assessed by mass spectrometry) could adequately account for the observed loss in macrophage content in plaques that undergo phenotypic regression. Instead, suppression of local proliferation of macrophages dominates phenotypic plaque regression in response to cholesterol lowering: the lower the levels of serum LDL-cholesterol and lipid contents in murine aortic and human carotid artery plaques, the lower the rates of in situ macrophage proliferation. Our study identifies macrophage proliferation as the predominant turnover determinant and an attractive target for inducing plaque regression.


Asunto(s)
Aterosclerosis/terapia , Atorvastatina/farmacología , Proliferación Celular/efectos de los fármacos , LDL-Colesterol/sangre , Dieta con Restricción de Grasas , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Macrófagos/efectos de los fármacos , Placa Aterosclerótica , Animales , Apolipoproteína E3/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , Proteínas de Transferencia de Ésteres de Colesterol/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Receptores de LDL/genética
17.
Trends Immunol ; 37(1): 32-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26748179

RESUMEN

Macrophages inhabit all major organs, and are capable of adapting their functions to meet the needs of their home tissues. The recent recognition that tissue macrophages derive from different sources, coupled with the notion that environmental cues and inflammatory stimuli can sculpt and agitate homeostasis, provides a frame of reference from which we can decipher the breadth and depth of macrophage activity. Here we discuss macrophages residing in the cardiovascular system, focusing particularly on their development and function in steady state and disease. Central to our discussion is the tension between macrophage ontogeny as a determinant of macrophage function, and the idea that tissues condition macrophage activities and supplant the influence of macrophage origins in favor of environmental demands.


Asunto(s)
Arterias/citología , Aterosclerosis/patología , Macrófagos/fisiología , Infarto del Miocardio/patología , Miocardio/citología , Animales , Diferenciación Celular , Desarrollo Embrionario , Humanos , Monocitos/fisiología
18.
Circ Res ; 121(4): 354-367, 2017 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-28637783

RESUMEN

RATIONALE: Inflammation is a key contributor to atherosclerosis. MicroRNA-146a (miR-146a) has been identified as a critical brake on proinflammatory nuclear factor κ light chain enhancer of activated B cells signaling in several cell types, including endothelial cells and bone marrow (BM)-derived cells. Importantly, miR-146a expression is elevated in human atherosclerotic plaques, and polymorphisms in the miR-146a precursor have been associated with risk of coronary artery disease. OBJECTIVE: To define the role of endogenous miR-146a during atherogenesis. METHODS AND RESULTS: Paradoxically, Ldlr-/- (low-density lipoprotein receptor null) mice deficient in miR-146a develop less atherosclerosis, despite having highly elevated levels of circulating proinflammatory cytokines. In contrast, cytokine levels are normalized in Ldlr-/-;miR-146a-/- mice receiving wild-type BM transplantation, and these mice have enhanced endothelial cell activation and elevated atherosclerotic plaque burden compared with Ldlr-/- mice receiving wild-type BM, demonstrating the atheroprotective role of miR-146a in the endothelium. We find that deficiency of miR-146a in BM-derived cells precipitates defects in hematopoietic stem cell function, contributing to extramedullary hematopoiesis, splenomegaly, BM failure, and decreased levels of circulating proatherogenic cells in mice fed an atherogenic diet. These hematopoietic phenotypes seem to be driven by unrestrained inflammatory signaling that leads to the expansion and eventual exhaustion of hematopoietic cells, and this occurs in the face of lower levels of circulating low-density lipoprotein cholesterol in mice lacking miR-146a in BM-derived cells. Furthermore, we identify sortilin-1(Sort1), a known regulator of circulating low-density lipoprotein levels in humans, as a novel target of miR-146a. CONCLUSIONS: Our study reveals that miR-146a regulates cholesterol metabolism and tempers chronic inflammatory responses to atherogenic diet by restraining proinflammatory signaling in endothelial cells and BM-derived cells.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , MicroARNs/metabolismo , Animales , Aterosclerosis/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Bovinos , VLDL-Colesterol/metabolismo , Dieta Aterogénica/efectos adversos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Receptores de LDL/metabolismo
20.
Clin Sci (Lond) ; 132(12): 1253-1256, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29930143

RESUMEN

The pathogenesis of thoracic aortic aneurysm and dissection (TAAD) is complex and incompletely understood. The hallmarks of the disease process are aortic inflammatory cell infiltration and protease mediated elastic fiber disruption. In a study recently published in Clinical Science (2018) 132 (6), 655-668), Liu et al explore the mechanism through which aortic vascular smooth cells and macrophages participate in TAAD using a mouse model. The authors propose that interleukin-3 (IL-3) released from aortic vascular smooth cells is central to the disease process. IL-3 stimulated matrix metalloproteinase 12 (MMP12) release from macrophages via mitogen activated protein kinase pathways. MMP12 is a protease known to be involved in both aortic aneurysm and dissection. IL-3 knockout mice had significantly reduced aortic wall MMP12, and reduced protease activity. This was associated with protection against TAAD.


Asunto(s)
Aneurisma de la Aorta Torácica , Animales , Interleucina-3 , Macrófagos , Metaloproteinasa 12 de la Matriz , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA