Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
1.
J Clin Invest ; 131(20)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34520398

RESUMEN

Tumor-infiltrating myeloid cells contribute to the development of the immunosuppressive tumor microenvironment. Myeloid cell expression of arginase 1 (ARG1) promotes a protumor phenotype by inhibiting T cell function and depleting extracellular l-arginine, but the mechanism underlying this expression, especially in breast cancer, is poorly understood. In breast cancer clinical samples and in our mouse models, we identified tumor-derived GM-CSF as the primary regulator of myeloid cell ARG1 expression and local immune suppression through a gene-KO screen of breast tumor cell-produced factors. The induction of myeloid cell ARG1 required GM-CSF and a low pH environment. GM-CSF signaling through STAT3 and p38 MAPK and acid signaling through cAMP were required to activate myeloid cell ARG1 expression in a STAT6-independent manner. Importantly, breast tumor cell-derived GM-CSF promoted tumor progression by inhibiting host antitumor immunity, driving a significant accumulation of ARG1-expressing myeloid cells compared with lung and melanoma tumors with minimal GM-CSF expression. Blockade of tumoral GM-CSF enhanced the efficacy of tumor-specific adoptive T cell therapy and immune checkpoint blockade. Taken together, we show that breast tumor cell-derived GM-CSF contributes to the development of the immunosuppressive breast cancer microenvironment by regulating myeloid cell ARG1 expression and can be targeted to enhance breast cancer immunotherapy.


Asunto(s)
Arginasa/fisiología , Neoplasias de la Mama/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Tolerancia Inmunológica , Células Mieloides/enzimología , Microambiente Tumoral , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , AMP Cíclico/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL
2.
Blood ; 137(26): 3591-3594, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33971000

RESUMEN

VEXAS syndrome (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) is a monogenic disease of adulthood caused by somatic mutations in UBA1 in hematopoietic progenitor cells. Patients develop inflammatory and hematologic symptoms. Myeloid-driven autoinflammation and progressive bone marrow failure lead to substantial morbidity and mortality. Effective medical treatments need to be identified. Reports in the current issue of Blood describe novel UBA1 genetic variants, treatment options, and insight into disease pathophysiology. VEXAS syndrome represents a prototype for a new class of diseases.


Asunto(s)
Genes Ligados a X , Enfermedades Genéticas Congénitas , Mutación , Trastornos Mieloproliferativos , Enzimas Activadoras de Ubiquitina/genética , Células Eritroides/enzimología , Enfermedades Genéticas Congénitas/diagnóstico por imagen , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/genética , Humanos , Masculino , Células Mieloides/enzimología , Trastornos Mieloproliferativos/diagnóstico por imagen , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Síndrome
3.
Front Immunol ; 12: 628156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34046031

RESUMEN

Brain myeloid cells, include infiltrating macrophages and resident microglia, play an essential role in responding to and inducing neurodegenerative diseases, such as Alzheimer's disease (AD). Genome-wide association studies (GWAS) implicate many AD casual and risk genes enriched in brain myeloid cells. Coordinated arginine metabolism through arginase 1 (Arg1) is critical for brain myeloid cells to perform biological functions, whereas dysregulated arginine metabolism disrupts them. Altered arginine metabolism is proposed as a new biomarker pathway for AD. We previously reported Arg1 deficiency in myeloid biased cells using lysozyme M (LysM) promoter-driven deletion worsened amyloidosis-related neuropathology and behavioral impairment. However, it remains unclear how Arg1 deficiency in these cells impacts the whole brain to promote amyloidosis. Herein, we aim to determine how Arg1 deficiency driven by LysM restriction during amyloidosis affects fundamental neurodegenerative pathways at the transcriptome level. By applying several bioinformatic tools and analyses, we found that amyloid-ß (Aß) stimulated transcriptomic signatures in autophagy-related pathways and myeloid cells' inflammatory response. At the same time, myeloid Arg1 deficiency during amyloidosis promoted gene signatures of lipid metabolism, myelination, and migration of myeloid cells. Focusing on Aß associated glial transcriptomic signatures, we found myeloid Arg1 deficiency up-regulated glial gene transcripts that positively correlated with Aß plaque burden. We also observed that Aß preferentially activated disease-associated microglial signatures to increase phagocytic response, whereas myeloid Arg1 deficiency selectively promoted homeostatic microglial signature that is non-phagocytic. These transcriptomic findings suggest a critical role for proper Arg1 function during normal and pathological challenges associated with amyloidosis. Furthermore, understanding pathways that govern Arg1 metabolism may provide new therapeutic opportunities to rebalance immune function and improve microglia/macrophage fitness.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Arginasa/metabolismo , Encéfalo/enzimología , Perfilación de la Expresión Génica , Microglía/enzimología , Células Mieloides/enzimología , Degeneración Nerviosa , Transcriptoma , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Arginasa/genética , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Redes Reguladoras de Genes , Haploinsuficiencia , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Mutación , Células Mieloides/patología
4.
Basic Res Cardiol ; 116(1): 31, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33929610

RESUMEN

Aircraft noise induces vascular and cerebral inflammation and oxidative stress causing hypertension and cardiovascular/cerebral dysfunction. With the present studies, we sought to determine the role of myeloid cells in the vascular vs. cerebral consequences of exposure to aircraft noise. Toxin-mediated ablation of lysozyme M+ (LysM+) myeloid cells was performed in LysMCreiDTR mice carrying a cre-inducible diphtheria toxin receptor. In the last 4d of toxin treatment, the animals were exposed to noise at maximum and mean sound pressure levels of 85 and 72 dB(A), respectively. Flow cytometry analysis revealed accumulation of CD45+, CD11b+, F4/80+, and Ly6G-Ly6C+ cells in the aortas of noise-exposed mice, which was prevented by LysM+ cell ablation in the periphery, whereas brain infiltrates were even exacerbated upon ablation. Aircraft noise-induced increases in blood pressure and endothelial dysfunction of the aorta and retinal/mesenteric arterioles were almost completely normalized by ablation. Correspondingly, reactive oxygen species in the aorta, heart, and retinal/mesenteric vessels were attenuated in ablated noise-exposed mice, while microglial activation and abundance in the brain was greatly increased. Expression of phagocytic NADPH oxidase (NOX-2) and vascular cell adhesion molecule-1 (VCAM-1) mRNA in the aorta was reduced, while NFκB signaling appeared to be activated in the brain upon ablation. In sum, we show dissociation of cerebral and peripheral inflammatory reactions in response to aircraft noise after LysM+ cell ablation, wherein peripheral myeloid inflammatory cells represent a dominant part of the pathomechanism for noise stress-induced cardiovascular effects and their central nervous counterparts, microglia, as key mediators in stress responses.


Asunto(s)
Arterias/enzimología , Encéfalo/enzimología , Encefalitis/prevención & control , Microglía/enzimología , Muramidasa/deficiencia , Células Mieloides/enzimología , Ruido del Transporte/efectos adversos , Enfermedades Vasculares Periféricas/prevención & control , Aeronaves , Animales , Arterias/fisiopatología , Encéfalo/patología , Modelos Animales de Enfermedad , Encefalitis/enzimología , Encefalitis/etiología , Encefalitis/patología , Eliminación de Gen , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Muramidasa/genética , Estrés Oxidativo , Enfermedades Vasculares Periféricas/enzimología , Enfermedades Vasculares Periféricas/etiología , Enfermedades Vasculares Periféricas/fisiopatología , Especies Reactivas de Oxígeno/metabolismo
6.
Int J Mol Sci ; 22(4)2021 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-33562184

RESUMEN

The deepest evolutionary branches of the trypsin/chymotrypsin family of serine proteases are represented by the digestive enzymes of the gastrointestinal tract and the multi-domain proteases of the blood coagulation and complement system. Similar to the very old digestive system, highly diverse cleavage specificities emerged in various cell lineages of the immune defense system during vertebrate evolution. The four neutrophil serine proteases (NSPs) expressed in the myelomonocyte lineage, neutrophil elastase, proteinase 3, cathepsin G, and neutrophil serine protease 4, collectively display a broad repertoire of (S1) specificities. The origin of NSPs can be traced back to a circulating liver-derived trypsin-like protease, the complement factor D ancestor, whose activity is tightly controlled by substrate-induced activation and TNFα-induced locally upregulated protein secretion. However, the present-day descendants are produced and converted to mature enzymes in precursor cells of the bone marrow and are safely sequestered in granules of circulating neutrophils. The potential site and duration of action of these cell-associated serine proteases are tightly controlled by the recruitment and activation of neutrophils, by stimulus-dependent regulated secretion of the granules, and by various soluble inhibitors in plasma, interstitial fluids, and in the inflammatory exudate. An extraordinary dynamic range and acceleration of immediate defense responses have been achieved by exploiting the high structural plasticity of the trypsin fold.


Asunto(s)
Linaje de la Célula , Monocitos/enzimología , Células Mieloides/enzimología , Serina Proteasas/metabolismo , Animales , Catepsina G/metabolismo , Humanos , Elastasa de Leucocito/metabolismo , Monocitos/citología , Mieloblastina/metabolismo , Células Mieloides/citología
7.
Basic Res Cardiol ; 116(1): 7, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523326

RESUMEN

A preclinical model of troponin I-induced myocarditis (AM) revealed a prominent role of the immunoproteasome (ip), the main immune cell-resident proteasome isoform, in heart-directed autoimmunity. Viral infection of the heart is a known trigger of cardiac autoimmunity, with the ip enhancing systemic inflammatory responses after infection with a cardiotropic coxsackievirusB3 (CV). Here, we used ip-deficient A/J-LMP7-/- mice to investigate the role of ip-mediated effects on adaptive immunity in CV-triggered myocarditis and found no alteration of the inflammatory heart tissue damage or cardiac function in comparison to wild-type controls. Aiming to define the impact of the systemic inflammatory storm under the control of ip proteolysis during CV infection, we targeted the ip in A/J mice with the inhibitor ONX 0914 after the first cycle of infection, when systemic inflammation has set in, well before cardiac inflammation. During established acute myocarditis, the ONX 0914 treatment group had the same reduction in cardiac output as the controls, with inflammatory responses in heart tissue being unaffected by the compound. Based on these findings and with regard to the known anti-inflammatory role of ONX 0914 in CV infection, we conclude that the efficacy of ip inhibitors for CV-triggered myocarditis in A/J mice relies on their immunomodulatory effects on the systemic inflammatory reaction.


Asunto(s)
Antiinflamatorios/farmacología , Infecciones por Coxsackievirus/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Células Mieloides/efectos de los fármacos , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Animales , Células Cultivadas , Infecciones por Coxsackievirus/enzimología , Infecciones por Coxsackievirus/inmunología , Modelos Animales de Enfermedad , Enterovirus Humano B/inmunología , Enterovirus Humano B/patogenicidad , Interacciones Huésped-Patógeno , Inflamación/enzimología , Inflamación/inmunología , Inflamación/virología , Masculino , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/inmunología , Células Mieloides/virología , Miocarditis/enzimología , Miocarditis/inmunología , Miocarditis/virología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/virología , Complejo de la Endopetidasa Proteasomal/genética , Proteolisis
8.
Cardiovasc Res ; 117(1): 162-177, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32077922

RESUMEN

AIMS: Heart failure (HF) ensuing myocardial infarction (MI) is characterized by the initiation of a systemic inflammatory response. We aimed to elucidate the impact of myelomonocytic cells and their activation by angiotensin II on vascular endothelial function in a mouse model of HF after MI. METHODS AND RESULTS: HF was induced in male C57BL/6J mice by permanent ligation of the left anterior descending coronary artery. Compared to sham, HF mice had significantly impaired endothelial function accompanied by enhanced mobilization of Sca-1+c-Kit+ haematopoietic stem cells and Sca-1-c-Kit+ common myeloid and granulocyte-macrophage progenitors in the bone marrow as well as increased vascular infiltration of CD11b+Ly6G-Ly6Chigh monocytes and accumulation of CD11b+ F4/80+ macrophages, assessed by flow cytometry. Using mice with Cre-inducible expression of diphtheria toxin receptor in myeloid cells, we selectively depleted lysozyme M+ myelomonocytic cells for 10 days starting 28 days after MI. While the cardiac phenotype remained unaltered until 38 days post-MI, myeloid cell depletion attenuated vascular accumulation of Nox2+CD45+ cells, endothelial dysfunction, oxidative stress, and vascular expression of adhesion molecules and angiotensin II receptor type 1 (AT1R). Pharmacological blockade of this receptor for 4 weeks did not significantly alter cardiac function, but mimicked the effects of myeloid cell depletion: telmisartan (20 mg/kg/day, fed to C57BL/6J mice) diminished bone marrow myelopoesis and myeloid reactive oxygen species production, attenuated endothelial leucocyte rolling and vascular accumulation of CD11b+Ly6G-Ly6Chigh monocytes and macrophages, resulting in improved vascular function with less abundance of Nox2+CD45+ cells. CONCLUSION: Endothelial dysfunction in HF ensuing MI is mediated by inflammatory Nox2+ myeloid cells infiltrating the vessel wall that can be targeted by AT1R blockade.


Asunto(s)
Angiotensina II/metabolismo , Células Endoteliales/metabolismo , Insuficiencia Cardíaca/etiología , Células Mieloides/enzimología , Infarto del Miocardio/complicaciones , NADPH Oxidasa 2/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Vasculitis/etiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/inmunología , Rodamiento de Leucocito , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Muramidasa/genética , Muramidasa/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Infarto del Miocardio/enzimología , Infarto del Miocardio/inmunología , Estrés Oxidativo , Transducción de Señal , Telmisartán/farmacología , Vasculitis/tratamiento farmacológico , Vasculitis/enzimología , Vasculitis/inmunología
9.
Oxid Med Cell Longev ; 2020: 7095902, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33312338

RESUMEN

The formation of reactive oxygen species (ROS) by the myeloid cell NADPH oxidase NOX2 is critical for the destruction of engulfed microorganisms. However, recent studies imply that ROS, formed by NOX2+ myeloid cells in the malignant microenvironment, exert multiple actions of relevance to the growth and spread of neoplastic cells. By generating ROS, tumor-infiltrating myeloid cells and NOX2+ leukemic myeloid cells may thus (i) compromise the function and viability of adjacent cytotoxic lymphocytes, including natural killer (NK) cells and T cells, (ii) oxidize DNA to trigger cancer-promoting somatic mutations, and (iii) affect the redox balance in cancer cells to control their proliferation and survival. Here, we discuss the impact of NOX2-derived ROS for tumorigenesis, tumor progression, regulation of antitumor immunity, and metastasis. We propose that NOX2 may be a targetable immune checkpoint in cancer.


Asunto(s)
Carcinogénesis , Leucemia , Mutación , NADPH Oxidasa 2 , Proteínas de Neoplasias , Especies Reactivas de Oxígeno , Microambiente Tumoral , Animales , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/metabolismo , Humanos , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , Leucemia/enzimología , Leucemia/genética , Leucemia/inmunología , Linfocitos Infiltrantes de Tumor/enzimología , Linfocitos Infiltrantes de Tumor/inmunología , Células Mieloides/enzimología , Células Mieloides/inmunología , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/inmunología , NADPH Oxidasa 2/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/enzimología , Linfocitos T/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
10.
Front Immunol ; 11: 938, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32499785

RESUMEN

Amino acid metabolism is a critical regulator of the immune response, and its modulating becomes a promising approach in various forms of immunotherapy. Insufficient concentrations of essential amino acids restrict T-cells activation and proliferation. However, only arginases, that degrade L-arginine, as well as enzymes that hydrolyze L-tryptophan are substantially increased in cancer. Two arginase isoforms, ARG1 and ARG2, have been found to be present in tumors and their increased activity usually correlates with more advanced disease and worse clinical prognosis. Nearly all types of myeloid cells were reported to produce arginases and the increased numbers of various populations of myeloid-derived suppressor cells and macrophages correlate with inferior clinical outcomes of cancer patients. Here, we describe the role of arginases produced by myeloid cells in regulating various populations of immune cells, discuss molecular mechanisms of immunoregulatory processes involving L-arginine metabolism and outline therapeutic approaches to mitigate the negative effects of arginases on antitumor immune response. Development of potent arginase inhibitors, with improved pharmacokinetic properties, may lead to the elaboration of novel therapeutic strategies based on targeting immunoregulatory pathways controlled by L-arginine degradation.


Asunto(s)
Arginasa/inmunología , Arginina/metabolismo , Células Mieloides/enzimología , Neoplasias/inmunología , Animales , Antineoplásicos/uso terapéutico , Arginasa/antagonistas & inhibidores , Ensayos Clínicos como Asunto , Humanos , Macrófagos/inmunología , Ratones , Células Progenitoras Mieloides/metabolismo , Neoplasias/tratamiento farmacológico
11.
Cell Death Dis ; 11(5): 305, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366830

RESUMEN

Death-associated protein kinase 1 (DAPK1, DAPk, DAPK) is known for its involvement in apoptosis and autophagy-associated cell death. Here, we identified an unexpected function of DAPK1 in suppressing necroptosis. DAPK1-deficiency renders macrophages and dendritic cells susceptible to necroptotic death. We also observed an inhibitory role for DAPK1 in necroptosis in HT-29 cells, since knockdown or knockout of DAPK1 in such cells increased their sensitivity to necroptosis. Increased necroptosis was associated with enhanced formation of the RIPK1-RIPK3-MLKL complex in these DAPK1-deficient cells. We further found that DAPK1-deficiency led to decreased MAPK activated kinase 2 (MK2) activation and reduced RIPK1 S321 phosphorylation, with this latter representing a critical step controlling necrosome formation. Most TNF signaling pathways, including ERK, JNK, and AKT, were not regulated by DAPK. In contrast, DAPK bound p38 MAPK and selectively promoted p38 MAPK activation, resulting in enhanced MK2 phosphorylation. Our results reveal a novel role for DAPK1 in inhibiting necroptosis and illustrate an unexpected selectivity for DAPK1 in promoting p38 MAPK-MK2 activation. Importantly, our study suggests that modulation of necroptosis and p38/MK2-mediated inflammation may be achieved by targeting DAPK1.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Necroptosis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Caspasa 8/metabolismo , Supervivencia Celular , Proteínas Quinasas Asociadas a Muerte Celular/deficiencia , Regulación hacia Abajo , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/patología , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Choque Séptico/metabolismo , Choque Séptico/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa
12.
J Immunol ; 204(8): 2088-2097, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32188756

RESUMEN

DNase 1-like 3 (DNase1L3), which belongs to DNase1 family, was originally identified as one of apoptosis- and necrosis-related endonucleases that fragmentate intranucleosomal DNA. A loss-of-function mutation has been reported in murine models of systemic lupus erythematosus (SLE) and in familial SLE patients. These reports suggest DNase1L3 plays an important role in the prevention of developing SLE; however, expression and function of DNase1L3 in human immune systems have been largely unclarified. As previous reports showed DNase1L3 is expressed in hematopoietic organs, we first analyzed expression levels of DNase1L3 in each subset of human peripheral blood cells by quantitative real-time PCR. Plasmacytoid dendritic cells showed the highest expression levels of DNase1L3 mRNA among peripheral blood cells. IL-4 enhanced DNase1L3 expression in monocytes, monocyte-derived dendritic cells, and monocyte-derived macrophages (MDMs), but not in T cells, B cells, or plasmacytoid dendritic cells. Together with IL-4, all-trans retinoic acid and apoptotic cells efficiently upregulated expression of DNalse1L3 in MDMs. As a result of intracellular signaling analysis, Jak1-IRS2-ERK/PI3K pathway was essential for IL-4-induced DNase1L3 expression. IL-4-treated monocyte-derived dendritic cells and MDMs secreted active DNase1L3 protein that could degrade liposome-DNA complexes, which were resistant to DNase1. Our results indicate DNase1L3 is secreted by innate immune cells and may play a critical role in the tissue homeostasis and on prevention of developing autoimmunity by degrading self-DNA.


Asunto(s)
Endodesoxirribonucleasas/biosíntesis , Homeostasis , Células Mieloides/enzimología , Células Cultivadas , ADN/inmunología , ADN/metabolismo , Endodesoxirribonucleasas/genética , Humanos
13.
Mol Neurobiol ; 57(5): 2447-2460, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32146679

RESUMEN

Microglial dysregulation, pertaining to impairment in phagocytosis, clearance and containment of amyloid-ß (Aß), and activation of neuroinflammation, has been posited to contribute to the pathogenesis of Alzheimer's disease (AD). Detailed cellular mechanisms that are disrupted during the disease course to display such impairment in microglia, however, remain largely undetermined. We hypothesize that loss of hematopoietic cell kinase (HCK), a phagocytosis-regulating member of the Src family tyrosine kinases that mediate signals from triggering receptor expressed on myeloid cells 2 and other immunoreceptors, impairs microglial homeostasis and Aß clearance, leading to the accelerated buildup of Aß pathology and cognitive decline during the early stage of neuropathological development. To elucidate the pivotal role of HCK in AD, we generated a constitutive knockout of HCK in the Tg2576 mouse model of AD. We found that HCK deficiency accelerated cognitive decline along with elevated Aß level and plaque burden, attenuated microglial Aß phagocytosis, induced iNOS expression in microglial clusters, and reduced pre-synaptic protein at the hippocampal regions. Our findings substantiate that HCK plays a prominent role in regulating microglial neuroprotective functions and attenuating early AD neuropathology.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Microglía/enzimología , Proteínas Proto-Oncogénicas c-hck/deficiencia , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Conducta Exploratoria , Femenino , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/patología , Prueba del Laberinto Acuático de Morris , Células Mieloides/enzimología , Neuroinmunomodulación , Fagocitosis , Placa Amiloide , Proteínas Proto-Oncogénicas c-hck/genética , Reconocimiento en Psicología
14.
Hepatology ; 72(4): 1191-1203, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31953865

RESUMEN

BACKGROUND AND AIMS: Indole is a microbiota metabolite that exerts anti-inflammatory responses. However, the relevance of indole to human non-alcoholic fatty liver disease (NAFLD) is not clear. It also remains largely unknown whether and how indole acts to protect against NAFLD. The present study sought to examine the association between the circulating levels of indole and liver fat content in human subjects and explore the mechanisms underlying indole actions in mice with diet-induced NAFLD. APPROACH AND RESULTS: In a cohort of 137 subjects, the circulating levels of indole were reversely correlated with body mass index. In addition, the circulating levels of indole in obese subjects were significantly lower than those in lean subjects and were accompanied with increased liver fat content. At the whole-animal level, treatment of high-fat diet (HFD)-fed C57BL/6J mice with indole caused significant decreases in the severity of hepatic steatosis and inflammation. In cultured cells, indole treatment stimulated the expression of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), a master regulatory gene of glycolysis, and suppressed macrophage proinflammatory activation in a PFKFB3-dependent manner. Moreover, myeloid cell-specific PFKFB3 disruption exacerbated the severity of HFD-induced hepatic steatosis and inflammation and blunted the effect of indole on alleviating diet-induced NAFLD phenotype. CONCLUSIONS: Taken together, our results demonstrate that indole is relevant to human NAFLD and capable of alleviating diet-induced NAFLD phenotypes in mice in a myeloid cell PFKFB3-dependent manner. Therefore, indole mimetic and/or macrophage-specific PFKFB3 activation may be the viable preventive and/or therapeutic approaches for inflammation-associated diseases including NAFLD.


Asunto(s)
Indoles/uso terapéutico , Inflamación/tratamiento farmacológico , Células Mieloides/enzimología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Fosfofructoquinasa-2/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Dieta Alta en Grasa , Femenino , Hepatocitos/metabolismo , Humanos , Indoles/sangre , Indoles/farmacología , Lipogénesis/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo
15.
Front Immunol ; 11: 604785, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613525

RESUMEN

c-Jun N-terminal protein kinase 1 (JNK1) is involved in multiple biological processes but its implication in inflammatory skin diseases is still poorly defined. Herein, we studied the role of JNK1 in the context of Aldara®-induced skin inflammation. We observed that constitutive ablation of JNK1 reduced Aldara®-induced acanthosis and expression of inflammatory markers. Conditional deletion of JNK1 in myeloid cells led to reduced skin inflammation, a finding that was associated with impaired Aldara®-induced inflammasome activation in vitro. Next, we evaluated the specific role of JNK1 in epidermal cells. We observed reduced Aldara®-induced acanthosis despite similar levels of inflammatory markers. Transcriptomic and epigenomic analysis of keratinocytes revealed the potential involvement of JNK1 in the EGFR signaling pathway. Finally, we show that inhibition of the EGFR pathway reduced Aldara®-induced acanthosis. Taken together, these data indicate that JNK1 plays a dual role in the context of psoriasis by regulating the production of inflammatory cytokines by myeloid cells and the sensitivity of keratinocytes to EGFR ligands. These results suggest that JNK1 could represent a valuable therapeutic target in the context of psoriasis.


Asunto(s)
Receptores ErbB/metabolismo , Queratinocitos/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Células Mieloides/enzimología , Psoriasis/enzimología , Piel/enzimología , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Epigenoma , Receptores ErbB/genética , Femenino , Imiquimod , Mediadores de Inflamación/metabolismo , Queratinocitos/inmunología , Queratinocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Células Mieloides/inmunología , Psoriasis/inducido químicamente , Psoriasis/inmunología , Psoriasis/patología , Transducción de Señal , Piel/inmunología , Piel/patología , Transcriptoma
16.
Biochem Biophys Res Commun ; 520(3): 573-579, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31615657

RESUMEN

BACKGROUND: Macrophages are ubiquitous in all stages of atherosclerosis, exerting tremendous impact on lesion progression and plaque stability. Because macrophages in atherosclerotic plaques express angiotensin-converting enzyme (ACE), current dogma posits that local myeloid-mediated effects worsen the disease. In contrast, we previously reported that myeloid ACE overexpression augments macrophage resistance to various immune challenges, including tumors, bacterial infection and Alzheimer's plaque deposition. Here, we sought to assess the impact of myeloid ACE on atherosclerosis. METHODS: A mouse model in which ACE is overexpressed in myelomonocytic lineage cells, called ACE10, was generated and sequentially crossed with ApoE-deficient mice to create ACE10/10ApoE-/- (ACE10/ApoE). Control mice were ACEWT/WTApoE-/- (WT/ApoE). Atherosclerosis was induced using an atherogenic diet alone, or in combination with unilateral nephrectomy plus deoxycorticosterone acetate (DOCA) salt for eight weeks. RESULTS: With an atherogenic diet alone or in combination with DOCA, the ACE10/ApoE mice showed significantly less atherosclerotic plaques compared to their WT/ApoE counterparts (p < 0.01). When recipient ApoE-/- mice were reconstituted with ACE10/10 bone marrow, these mice showed significantly reduced lesion areas compared to recipients reconstituted with wild type bone marrow. Furthermore, transfer of ACE-deficient bone marrow had no impact on lesion area. CONCLUSION: Our data indicate that while myeloid ACE may not be required for atherosclerosis, enhanced ACE expression paradoxically reduced disease progression.


Asunto(s)
Aterosclerosis/enzimología , Aterosclerosis/prevención & control , Células Mieloides/enzimología , Peptidil-Dipeptidasa A/metabolismo , Animales , Aterosclerosis/genética , Presión Sanguínea , Trasplante de Médula Ósea , Linaje de la Célula/genética , Colesterol/sangre , Dieta Aterogénica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Células Mieloides/patología , Peptidil-Dipeptidasa A/genética , Regulación hacia Arriba
17.
J Biol Chem ; 294(43): 15836-15849, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31495784

RESUMEN

Cholesterol esters are a key ingredient of foamy cells in atherosclerotic lesions; their formation is catalyzed by two enzymes: acyl-CoA:cholesterol acyltransferases (ACATs; also called sterol O-acyltransferases, or SOATs) ACAT1 and ACAT2. ACAT1 is present in all body cells and is the major isoenzyme in macrophages. Whether blocking ACAT1 benefits atherosclerosis has been under debate for more than a decade. Previously, our laboratory developed a myeloid-specific Acat1 knockout (KO) mouse (Acat1-M/-M), devoid of ACAT1 only in macrophages, microglia, and neutrophils. In previous work using the ApoE KO (ApoE-/-) mouse model for early lesions, Acat1-M/-M significantly reduced lesion macrophage content and suppressed atherosclerosis progression. In advanced lesions, cholesterol crystals become a prominent feature. Here we evaluated the effects of Acat1-M/-M in the ApoE KO mouse model for more advanced lesions and found that mice lacking myeloid Acat1 had significantly reduced lesion cholesterol crystal contents. Acat1-M/-M also significantly reduced lesion size and macrophage content without increasing apoptotic cell death. Cell culture studies showed that inhibiting ACAT1 in macrophages caused cells to produce less proinflammatory responses upon cholesterol loading by acetyl low-density lipoprotein. In advanced lesions, Acat1-M/-M reduced but did not eliminate foamy cells. In advanced plaques isolated from ApoE-/- mice, immunostainings showed that both ACAT1 and ACAT2 are present. In cell culture, both enzymes are present in macrophages and smooth muscle cells and contribute to cholesterol ester biosynthesis. Overall, our results support the notion that targeting ACAT1 or targeting both ACAT1 and ACAT2 in macrophages is a novel strategy to treat advanced lesions.


Asunto(s)
Aterosclerosis/enzimología , Aterosclerosis/prevención & control , Inflamación/patología , Macrófagos Peritoneales/enzimología , Células Mieloides/enzimología , Esterol O-Aciltransferasa/metabolismo , Animales , Apolipoproteínas E , Apoptosis , Aterosclerosis/patología , Colesterol/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Silenciador del Gen , Hidroxicolesteroles/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Células Mieloides/patología , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células RAW 264.7
18.
J Am Soc Nephrol ; 30(9): 1674-1685, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315922

RESUMEN

BACKGROUND: Following an acute insult, macrophages regulate renal fibrogenesis through the release of various factors that either encourage the synthesis of extracellular matrix synthesis or the degradation of matrix via endocytosis, proteolysis, or both. However, the roles of infiltrating versus resident myeloid cells in these opposing processes require elucidation. The transcription factor Twist1 controls diverse essential cellular functions through induction of several downstream targets, including matrix metalloproteinases (MMPs). In macrophages, Twist1 can influence patterns of cytokine generation, but the role of macrophage Twist1 in renal fibrogenesis remains undefined. METHODS: To study Twist1 functions in different macrophage subsets during kidney scar formation, we used two conditional mutant mouse models in which Twist1 was selectively ablated either in infiltrating, inflammatory macrophages or in resident tissue macrophages. We assessed fibrosis-related parameters, matrix metallopeptidase 13 (MMP13, or collagen 3, which catalyzes collagen degradation), inflammatory cytokines, and other factors in these Twist1-deficient mice compared with wild-type controls after subjecting the animals to unilateral ureteral obstruction. We also treated wild-type and Twist1-deficient mice with an MMP13 inhibitor after unilateral ureteral obstruction. RESULTS: Twist1 in infiltrating inflammatory macrophages but not in resident macrophages limited kidney fibrosis after ureteral obstruction by driving extracellular matrix degradation. Moreover, deletion of Twist1 in infiltrating macrophages attenuated the expression of MMP13 in CD11b+Ly6Clo myeloid cells. Inhibition of MMP13 abrogated the protection from renal fibrosis afforded by macrophage Twist1. CONCLUSIONS: Twist1 in infiltrating myeloid cells mitigates interstitial matrix accumulation in the injured kidney by promoting MMP13 production, which drives extracellular matrix degradation. These data highlight the complex cell-specific actions of Twist1 in the pathogenesis of kidney fibrosis.


Asunto(s)
Matriz Extracelular/metabolismo , Enfermedades Renales/metabolismo , Riñón/metabolismo , Riñón/patología , Macrófagos/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Actinas/metabolismo , Animales , Benzofuranos/farmacología , Receptor 1 de Quimiocinas CX3C/metabolismo , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Expresión Génica , Hidroxiprolina/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/patología , Macrófagos Peritoneales/metabolismo , Masculino , Metaloproteinasa 13 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Morfolinas/farmacología , Células Mieloides/enzimología , Proteína 1 Relacionada con Twist/genética , Obstrucción Ureteral/complicaciones
19.
Am J Physiol Heart Circ Physiol ; 317(2): H364-H374, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31149833

RESUMEN

Reduced vasodilator properties of insulin in obesity are caused by changes in perivascular adipose tissue and contribute to microvascular dysfunction in skeletal muscle. The causes of this dysfunction are unknown. The effects of a short-term Western diet on JNK2-expressing cells in perivascular adipose tissue (PVAT) on insulin-induced vasodilation and perfusion of skeletal muscle were assessed. In vivo, 2 wk of Western diet (WD) reduced whole body insulin sensitivity and insulin-stimulated muscle perfusion, determined using contrast ultrasonography during the hyperinsulinemic clamp. Ex vivo, WD triggered accumulation of PVAT in skeletal muscle and blunted its ability to facilitate insulin-induced vasodilation. Labeling of myeloid cells with green fluorescent protein identified bone marrow as a source of PVAT in muscle. To study whether JNK2-expressing inflammatory cells from bone marrow were involved, we transplanted JNK2-/- bone marrow to WT mice. Deletion of JNK2 in bone marrow rescued the vasodilator phenotype of PVAT during WD exposure. JNK2 deletion in myeloid cells prevented the WD-induced increase in F4/80 expression. Even though WD and JNK2 deletion resulted in specific changes in gene expression of PVAT; epididymal and subcutaneous adipose tissue; expression of tumor necrosis factor-α, interleukin-1ß, interleukin-6, or protein inhibitor of STAT1 was not affected. In conclusion, short-term Western diet triggers infiltration of JNK2-positive myeloid cells into PVAT, resulting in PVAT dysfunction, nonclassical inflammation, and loss of insulin-induced vasodilatation in vivo and ex vivo.NEW & NOTEWORTHY We demonstrate that in the earliest phase of weight gain, changes in perivascular adipose tissue in muscle impair insulin-stimulated muscle perfusion. The hallmark of these changes is infiltration by inflammatory cells. Deletion of JNK2 from the bone marrow restores the function of perivascular adipose tissue to enhance insulin's vasodilator effects in muscle, showing that the bone marrow contributes to regulation of muscle perfusion.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Resistencia a la Insulina , Insulina/farmacología , Microvasos/efectos de los fármacos , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Músculo Esquelético/irrigación sanguínea , Células Mieloides/enzimología , Obesidad/enzimología , Vasodilatación/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Trasplante de Médula Ósea , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/fisiopatología , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Obesidad/etiología , Obesidad/fisiopatología , Flujo Sanguíneo Regional , Factores de Tiempo , Aumento de Peso
20.
J Exp Med ; 216(7): 1700-1723, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31126966

RESUMEN

The RNase Regnase-1 is a master RNA regulator in macrophages and T cells that degrades cellular and viral RNA upon NF-κB signaling. The roles of its family members, however, remain largely unknown. Here, we analyzed Regnase-3-deficient mice, which develop hypertrophic lymph nodes. We used various mice with immune cell-specific deletions of Regnase-3 to demonstrate that Regnase-3 acts specifically within myeloid cells. Regnase-3 deficiency systemically increased IFN signaling, which increased the proportion of immature B and innate immune cells, and suppressed follicle and germinal center formation. Expression analysis revealed that Regnase-3 and Regnase-1 share protein degradation pathways. Unlike Regnase-1, Regnase-3 expression is high specifically in macrophages and is transcriptionally controlled by IFN signaling. Although direct targets in macrophages remain unknown, Regnase-3 can bind, degrade, and regulate mRNAs, such as Zc3h12a (Regnase-1), in vitro. These data indicate that Regnase-3, like Regnase-1, is an RNase essential for immune homeostasis but has diverged as key regulator in the IFN pathway in macrophages.


Asunto(s)
Homeostasis/inmunología , Inmunidad Innata , Interferones/metabolismo , Células Mieloides/metabolismo , Ribonucleasas/metabolismo , Regiones no Traducidas 3' , Animales , Autoinmunidad , Linfocitos B/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribonucleasas/genética , Transducción de Señal , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...