Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.284
Filtrar
1.
Nat Commun ; 15(1): 8663, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375351

RESUMEN

Listeria monocytogenes (LM) possesses the ability to breach multiple barriers and elicit intricate immune responses. However, there remains a lack of explicit understanding regarding how LM evades innate immune surveillance within the body. Here, we utilized liver intravital imaging to elucidate the dynamic process of LM during infection in the liver. We discovered that LM can rapidly escape from Kupffer cells (KCs) through listeriolysin O (LLO) and proliferate within hepatocytes. Upon LM exposure to the hepatic sinusoids, neutrophils rapidly aggregate at the site of infection. Subsequently, LM can induce type I interferon (IFN-I) production primarily in the spleen, which acts systemically on neutrophils to hamper their swarming by deactivating the ERK pathway, thus evading neutrophil-mediated eradication. Furthermore, our findings suggest that virus-induced IFN-I suppresses neutrophil swarming, and COVID-19 patients exhibit impaired neutrophil aggregation function. In conclusion, our findings provide compelling evidence demonstrating that intracellular bacteria represented by LM can hijack host defense mechanisms against viral infections to evade immune surveillance. Additionally, impaired neutrophil swarming caused by IFN-I is one of the significant factors contributing to the increased susceptibility to bacterial infections following viral infections.


Asunto(s)
COVID-19 , Interferón Tipo I , Macrófagos del Hígado , Listeria monocytogenes , Listeriosis , Neutrófilos , Animales , Femenino , Humanos , Masculino , Ratones , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/inmunología , COVID-19/inmunología , COVID-19/virología , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Hepatocitos/virología , Hepatocitos/inmunología , Evasión Inmune , Inmunidad Innata , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Macrófagos del Hígado/inmunología , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Listeriosis/microbiología , Hígado/inmunología , Hígado/virología , Hígado/microbiología , Sistema de Señalización de MAP Quinasas/inmunología , Ratones Endogámicos C57BL , Neutrófilos/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Bazo/inmunología
2.
Nat Commun ; 15(1): 8690, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375367

RESUMEN

Primary biliary cholangitis (PBC) is a chronic autoimmune liver disease characterized by multilineage immune dysregulation, which subsequently causes inflammation, fibrosis, and even cirrhosis of liver. Due to the limitation of traditional assays, the local hepatic immunopathogenesis of PBC has not been fully characterized. Here, we utilize single-cell RNA sequencing technology to depict the immune cell landscape and decipher the molecular mechanisms of PBC patients. We reveal that cholangiocytes and hepatic stellate cells are involved in liver inflammation and fibrosis. Moreover, Kupffer cells show increased levels of inflammatory factors and decreased scavenger function related genes, while T cells exhibit enhanced levels of inflammatory factors and reduced cytotoxicity related genes. Interestingly, we identify a liver-resident Th1-like population with JAK-STAT activation in the livers of both PBC patients and murine PBC model. Finally, blocking the JAK-STAT pathway alleviates the liver inflammation and eliminates the liver-resident Th1-like cells in the murine PBC model. In conclusion, our comprehensive single-cell transcriptome profiling expands the understanding of pathological mechanisms of PBC and provides potential targets for the treatment of PBC in patients.


Asunto(s)
Quinasas Janus , Cirrosis Hepática Biliar , Hígado , Factores de Transcripción STAT , Análisis de la Célula Individual , Células TH1 , Animales , Análisis de la Célula Individual/métodos , Células TH1/inmunología , Humanos , Hígado/patología , Hígado/metabolismo , Hígado/inmunología , Ratones , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/inmunología , Cirrosis Hepática Biliar/patología , Cirrosis Hepática Biliar/metabolismo , Factores de Transcripción STAT/metabolismo , Factores de Transcripción STAT/genética , Quinasas Janus/metabolismo , Quinasas Janus/genética , Modelos Animales de Enfermedad , Análisis de Secuencia de ARN/métodos , Ratones Endogámicos C57BL , Femenino , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/inmunología , Células Estrelladas Hepáticas/patología , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/inmunología , Inflamación/genética , Inflamación/patología , Inflamación/metabolismo , Transducción de Señal , Transcriptoma , Perfilación de la Expresión Génica , Masculino
3.
Results Probl Cell Differ ; 74: 175-209, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39406906

RESUMEN

The liver fulfils a plethora of metabolic and immunological functions. Liver macrophages are a heterogeneous immune cell population with high plasticity and are important for maintaining normal liver function but are also critically involved in disease processes. In this chapter, we review the heterogeneity and multifaceted functions of hepatic macrophages in liver health and in disease conditions, including acute liver injury, chronic liver diseases, and hepatocellular carcinoma. Under homeostatic conditions, the tissue resident Kupffer cells are phagocytic cells that have important functions in immune surveillance, antigen presentation, and metabolic regulation while the roles of other populations such as capsular, peritoneal, or monocyte-derived macrophages in liver health are less clearly defined. Upon liver injury, Kupffer cell numbers are markedly reduced while monocyte-derived macrophages significantly expand and take critical roles in driving and resolving liver injury, including important pathogenic involvements in inflammation, fibrosis, and regeneration. They also create and maintain an immunosuppressive and immune-excluded microenvironment in hepatocellular carcinoma. Single-cell and spatial omics technologies are significantly expanding our understanding of the diversity and plasticity of macrophage populations under different conditions and enable the reliable identification of specific hepatic macrophage subsets. This knowledge can now be applied to dissect the exact contributions of distinct macrophage populations to disease processes and hopefully will pave the way for new therapeutic interventions.


Asunto(s)
Hepatopatías , Hígado , Macrófagos , Humanos , Animales , Hígado/inmunología , Hígado/patología , Macrófagos/inmunología , Hepatopatías/inmunología , Hepatopatías/patología , Macrófagos del Hígado/inmunología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología
4.
Nat Commun ; 15(1): 8341, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333539

RESUMEN

Hypercholesterolemia is a major risk factor for atherosclerosis and associated cardiovascular diseases. The liver plays a key role in the regulation of plasma cholesterol levels and hosts a large population of tissue-resident macrophages known as Kupffer cells (KCs). KCs are located in the hepatic sinusoids where they ensure key functions including blood immune surveillance. However, how KCs homeostasis is affected by the build-up of cholesterol-rich lipoproteins that occurs in the circulation during hypercholesterolemia remains unknown. Here, we show that embryo-derived KCs (EmKCs) accumulate large amounts of lipoprotein-derived cholesterol, in part through the scavenger receptor CD36, and massively expand early after the induction of hypercholesterolemia. After this rapid adaptive response, EmKCs exhibit mitochondrial oxidative stress and their numbers gradually diminish while monocyte-derived KCs (MoKCs) with reduced cholesterol-loading capacities seed the KC pool. Decreased proportion of EmKCs in the KC pool enhances liver cholesterol content and exacerbates hypercholesterolemia, leading to accelerated atherosclerotic plaque development. Together, our data reveal that KC homeostasis is perturbed during hypercholesterolemia, which in turn alters the control of plasma cholesterol levels and increases atherosclerosis.


Asunto(s)
Aterosclerosis , Antígenos CD36 , Colesterol , Hipercolesterolemia , Macrófagos del Hígado , Hígado , Ratones Endogámicos C57BL , Macrófagos del Hígado/metabolismo , Animales , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Colesterol/metabolismo , Colesterol/sangre , Hígado/metabolismo , Hígado/patología , Ratones , Antígenos CD36/metabolismo , Antígenos CD36/genética , Masculino , Monocitos/metabolismo , Estrés Oxidativo , Placa Aterosclerótica/patología , Placa Aterosclerótica/metabolismo , Ratones Noqueados , Femenino , Homeostasis
5.
Phytomedicine ; 134: 155561, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39217654

RESUMEN

BACKGROUND: Didymin is a dietary flavonoid originally discovered by our group as a potent anti-ulcerative colitis (UC) agent. However, whether didymin plays a protective role in UC-associated inflammatory liver injury is still unclear. PURPOSE: This study aimed to evaluate the therapeutic potential of didymin on UC-associated inflammatory liver injury and explore the underlying mechanism. STUDY DESIGN AND METHODS: Colitis model was established in C57BL/6 mice by exposure to DSS, and didymin was administrated intragastrically for consecutive 10 days. The inflammatory liver injury was assessed by levels of alanine aminotransferase (ALT) and aspartate transaminase (AST) in serum and histopathological damage in the liver. In vitro Kupffer cells and RAW264.7 cells challenged with lipopolysaccharides (LPS) were used to explore the modulatory activity of didymin on pro-inflammatory cytokines secretion and Notch1 signaling pathway activation. RESULTS: Didymin significantly mitigated liver coefficiency, ALT and AST levels in serum, and the hepatic histopathological damage caused by DSS-induced acute and chronic colitis. The mRNA expressions of pro-inflammatory factors including Tnf, Il1, and Il6 in liver tissues, Kupffer cells, and RAW264.7 cells stimulated by the influx of LPS was significantly deprived after didymin treatment. Mechanistically, didymin obstructed the protein expression, nuclear translocation of notch intracellular domain 1 (Notch1-ICD) and mRNA expression of hairy and enhancer of split 1 (Hes1). Further, the inhibitory mechanism of the Notch1-Hes1 pathway was dependent on c-Cbl-mediated Notch1-ICD lysosomal degradation. CONCLUSION: Our study verified for the first time that didymin could prevent UC-associated diseases, such as inflammatory liver injury, and the mechanism was related to facilitating Notch1 lysosomal degradation rather than proteasome degradation via promoting protein expression of c-Cbl in macrophages. Our findings that the inhibition of Notch1 signaling transduction helps to alleviate UC-associated liver injury provides possible therapeutics for the treatment of colitis and also furnishes a research paradigm for the study of flavonoids with similar structures.


Asunto(s)
Colitis Ulcerosa , Hígado , Receptor Notch1 , Animales , Masculino , Ratones , Alanina Transaminasa/sangre , Aspartato Aminotransferasas/sangre , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Citocinas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Flavonoides/farmacología , Glicósidos , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Lipopolisacáridos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones Endogámicos C57BL , Células RAW 264.7 , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Nat Commun ; 15(1): 8388, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333125

RESUMEN

Methionine adenosyltransferase 2 A (MAT2A) and MAT2B are essential for hepatic stellate cells (HSCs) activation. Forkhead box M1 (FOXM1) transgenic mice develop liver inflammation and fibrosis. Here we examine if they crosstalk in male mice. We found FOXM1/MAT2A/2B are upregulated after bile duct ligation (BDL) and carbon tetrachloride (CCl4) treatment in hepatocytes, HSCs and Kupffer cells (KCs). FDI-6, a FOXM1 inhibitor, attenuates the development and reverses the progression of CCl4-induced fibrosis while lowering the expression of FOXM1/MAT2A/2B, which exert reciprocal positive regulation on each other transcriptionally. Knocking down any of them lowers HSCs and KCs activation. Deletion of FOXM1 in hepatocytes, HSCs, and KCs protects from BDL-mediated inflammation and fibrosis comparably. Interestingly, HSCs from Foxm1Hep-/-, hepatocytes from Foxm1HSC-/-, and HSCs and hepatocytes from Foxm1KC-/- have lower FOXM1/MAT2A/2B after BDL. This may be partly due to transfer of extracellular vesicles between different cell types. Altogether, FOXM1/MAT2A/MAT2B axis drives liver inflammation and fibrosis.


Asunto(s)
Tetracloruro de Carbono , Proteína Forkhead Box M1 , Células Estrelladas Hepáticas , Hepatocitos , Macrófagos del Hígado , Cirrosis Hepática , Metionina Adenosiltransferasa , Animales , Metionina Adenosiltransferasa/metabolismo , Metionina Adenosiltransferasa/genética , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/genética , Masculino , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Ratones , Hepatocitos/metabolismo , Hepatocitos/patología , Macrófagos del Hígado/metabolismo , Tetracloruro de Carbono/toxicidad , Células Estrelladas Hepáticas/metabolismo , Ratones Endogámicos C57BL , Hígado/patología , Hígado/metabolismo , Ratones Noqueados , Ratones Transgénicos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Humanos , Conductos Biliares/patología , Conductos Biliares/metabolismo , Conductos Biliares/cirugía
7.
Immunity ; 57(10): 2310-2327.e6, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39317200

RESUMEN

The liver macrophage population comprises resident Kupffer cells (KCs) and monocyte-derived macrophages with distinct pro- or anti-inflammatory properties that affect the severity and course of liver diseases. The mechanisms underlying macrophage differentiation and functions in metabolic dysfunction-associated steatotic liver disease and/or steatohepatitis (MASLD/MASH) remain mostly unknown. Using single-cell RNA sequencing (scRNA-seq) and fate mapping of hepatic macrophage subpopulations, we unraveled the temporal and spatial dynamics of distinct monocyte and monocyte-derived macrophage subsets in MASH. We revealed a crucial role for the Notch-Recombination signal binding protein for immunoglobulin kappa J region (RBPJ) signaling pathway in controlling the monocyte-to-macrophage transition, with Rbpj deficiency blunting inflammatory macrophages and monocyte-derived KC differentiation and conversely promoting the emergence of protective Ly6Clo monocytes. Mechanistically, Rbpj deficiency promoted lipid uptake driven by elevated CD36 expression in Ly6Clo monocytes, enhancing their protective interactions with endothelial cells. Our findings uncover the crucial role of Notch-RBPJ signaling in monocyte-to-macrophage transition and will aid in the design of therapeutic strategies for MASH treatment.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Inflamación , Macrófagos , Receptores Notch , Transducción de Señal , Animales , Receptores Notch/metabolismo , Ratones , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Hígado Graso/metabolismo , Hígado Graso/inmunología , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Diferenciación Celular , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/inmunología , Ratones Noqueados , Humanos , Hígado/metabolismo , Hígado/patología
8.
Ecotoxicol Environ Saf ; 285: 117050, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39278002

RESUMEN

Trichloroethylene (TCE) is a common environmental contaminant that can induce occupational dermatitis medicamentosa-like TCE (ODMLT), where the liver damage is the most common complication. The study aims to uncover the underlying mechanism of TCE-sensitization-induced liver damage by targeting specific exosomal microRNAs (miRNAs). Among the enriched serum exosomal miRNAs of ODMLT patients, miR-205-5p had a significant correlation coefficient with the liver function damage indicators. Moreover, retinoic acid receptor-related orphan receptor α (RORα) was identified as a direct target of miR-205-5p via specific binding. Further experiments showed that kupffer cells (KCs) underwent M1 phenotypic and functional changes in liver injury induced by TCE which were alleviated by reducing the expression of miR-205-5p. However, this alleviation was reversed by the RORα antagonist SR1001. In vitro experiments showed that miR-205-5p promoted M1 polarization of macrophages and enhanced the secretion of inflammatory factors by regulating RORα. An increase in RORα reversed the polarization direction of M1-type macrophages and reduced the secretion of proinflammatory factors. In addition, pretreatment of mice with SR1078, a specific RORα agonist, effectively blocked M1 polarization of KCs and reduced the severity of TCE-induced liver injury. Our study uncovers that miR-205-5p regulates KC M1 polarization by targeting RORα in immune liver injury induced by TCE sensitization, providing new insight into the molecular mechanisms and new therapeutic targets for ODMLT.


Asunto(s)
Macrófagos del Hígado , MicroARNs , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Tricloroetileno , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , MicroARNs/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Tricloroetileno/toxicidad , Animales , Ratones , Humanos , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Exosomas/metabolismo , Masculino , Ratones Endogámicos C57BL
9.
Int J Mol Sci ; 25(18)2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39337381

RESUMEN

Both DNA 5-methylcytosine (5mC) and RNA N6-methyladenosine (m6A) modifications are reported to participate in cellular stress responses including inflammation. Phosphoenolpyruvate carboxykinase 2 (PCK2) is upregulated in Kupffer cells (KCs) to facilitate the proinflammatory phosphorylation signaling cascades upon LPS stimulation, yet the role of 5mC and m6A in PCK2 upregulation remain elusive. Here, we report that the significantly augmented PCK2 mRNA and protein levels are associated with global 5mC demethylation coupled with m6A hypermethylation in LPS-activated KCs. The suppression of 5mC demethylation or m6A hypermethylation significantly alleviates the upregulation of PCK2 and proinflammatory cytokines in LPS-challenged KCs. Further reciprocal tests indicate 5mC demethylation is upstream of m6A hypermethylation. Specifically, CpG islands in the promoters of PCK2 and RNA methyltransferase (METTL3 and METTL14) genes are demethylated, while the 3'UTR of PCK2 mRNA is m6A hypermethylated, in LPS-stimulated KCs. These modifications contribute to the transactivation of the PCK2 gene as well as increased PCK2 mRNA stability and protein production via a m6A-mediated mechanism with IGF2BP1 as the reader protein. These results indicate that DNA 5mC and RNA m6A collaborate to upregulate PCK2 expression, respectively, at the transcriptional and post-transcriptional levels during KC activation.


Asunto(s)
5-Metilcitosina , Adenosina , Metilación de ADN , Macrófagos del Hígado , Regulación hacia Arriba , Animales , Ratones , 5-Metilcitosina/metabolismo , 5-Metilcitosina/análogos & derivados , Adenosina/análogos & derivados , Adenosina/metabolismo , Islas de CpG , Macrófagos del Hígado/metabolismo , Lipopolisacáridos/farmacología , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , ARN Mensajero/genética
10.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(4): 603-609, 2024 Aug.
Artículo en Chino | MEDLINE | ID: mdl-39223025

RESUMEN

Kupffer cells (KC),an important subset of immune cells in the liver,are essential for maintaining tissue homeostasis and responding quickly to liver damage.The complement receptor of the immunoglobulin superfamily (CRIg) is a receptor protein on the KC membrane.CRIg can not only capture pathogens in the blood flowing through the liver by complement binding but also mediate immune responses by regulating immune cells in the liver.Recent studies have confirmed the role of CRIg in regulating liver immunity.This article reviews the main modes of action of CRIg and the research progress of CRIg in regulating liver immunity.


Asunto(s)
Macrófagos del Hígado , Hígado , Receptores de Complemento , Humanos , Hígado/inmunología , Hígado/metabolismo , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Animales
11.
Sci Transl Med ; 16(764): eadi0284, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39259813

RESUMEN

Proinflammatory hepatic macrophage activation plays a key role in the development of nonalcoholic steatohepatitis (NASH). This involves increased embryonic hepatic Kupffer cell (KC) death, facilitating the replacement of KCs with bone marrow-derived recruited hepatic macrophages (RHMs) that highly express proinflammatory genes. Moreover, phago/efferocytic activity of KCs is diminished in NASH, enhancing liver inflammation. However, the molecular mechanisms underlying these changes in KCs are not known. Here, we show that hypoxia-inducible factor 2α (HIF-2α) mediates NASH-associated decreased KC growth and efferocytosis by enhancing lysosomal stress. At the molecular level, HIF-2α stimulated mammalian target of rapamycin (mTOR)- and extracellular signal-regulated kinase-dependent inhibitory transcription factor EB (TFEB) phosphorylation, leading to decreased lysosomal and phagocytic gene expression. With increased metabolic stress and phago/efferocytic burden in NASH, these changes were sufficient to increase lysosomal stress, causing decreased efferocytosis and lysosomal cell death. Of interest, HIF-2α-dependent TFEB regulation only occurred in KCs but not RHMs. Instead, in RHMs, HIF-2α promoted mitochondrial reactive oxygen species production and proinflammatory activation by increasing ANT2 expression and mitochondrial permeability transition. Consequently, myeloid lineage-specific or KC-specific HIF-2α depletion or the inhibition of mTOR-dependent TFEB inhibition using antisense oligonucleotide treatment protected against the development of NASH in mice. Moreover, treatment with an HIF-2α-specific inhibitor reduced inflammatory and fibrogenic gene expression in human liver spheroids cultured under a NASH-like condition. Together, our results suggest that macrophage subtype-specific effects of HIF-2α collectively contribute to the proinflammatory activation of liver macrophages, leading to the development of NASH.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Macrófagos del Hígado , Hígado , Activación de Macrófagos , Enfermedad del Hígado Graso no Alcohólico , Macrófagos del Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Muerte Celular , Lisosomas/metabolismo , Fagocitosis , Humanos , Especies Reactivas de Oxígeno/metabolismo , Inflamación/patología , Inflamación/metabolismo , Ratones Endogámicos C57BL , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo
12.
Int J Mol Sci ; 25(17)2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39273394

RESUMEN

Purinergic signaling has emerged as an important paracrine-autocrine intercellular system that regulates physiological and pathological processes in practically all organs of the body. Although this system has been thoroughly defined since the nineties, recent research has made substantial advances regarding its role in aspects of liver physiology. However, most studies have mainly targeted the entire organ, 70% of which is made up of parenchymal cells or hepatocytes. Because of its physiological role, the liver is exposed to toxic metabolites, such as xenobiotics, drugs, and fatty acids, as well as to pathogens such as viruses and bacteria. Under injury conditions, all cell types within the liver undergo adaptive changes. In this context, the concentration of extracellular ATP has the potential to increase dramatically. Indeed, this purinergic response has not been studied in sufficient detail in non-parenchymal liver cells. In the present review, we systematize the physiopathological adaptations related to the purinergic system in chronic liver diseases of non-parenchymal liver cells, such as hepatic stellate cells, Kupffer cells, sinusoidal endothelial cells, and cholangiocytes. The role played by non-parenchymal liver cells in these circumstances will undoubtedly be strategic in understanding the regenerative activities that support the viability of this organ under stressful conditions.


Asunto(s)
Hígado , Receptores Purinérgicos , Transducción de Señal , Humanos , Animales , Hígado/metabolismo , Receptores Purinérgicos/metabolismo , Macrófagos del Hígado/metabolismo , Células Estrelladas Hepáticas/metabolismo , Adenosina Trifosfato/metabolismo , Hepatopatías/metabolismo , Hepatopatías/patología , Hepatocitos/metabolismo
13.
Redox Biol ; 76: 103314, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39163766

RESUMEN

Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive form of nonalcoholic fatty liver disease characterised by fat accumulation, inflammation, oxidative stress, fibrosis, and impaired liver regeneration. In this study, we found that heme oxygenase-1 (HO-1) is induced in both MASH patients and in a MASH mouse model. Further, hepatic carbon monoxide (CO) levels in MASH model mice were >2-fold higher than in healthy mice, suggesting that liver HO-1 is activated as MASH progresses. Based on these findings, we used CO-loaded red blood cells (CO-RBCs) as a CO donor in the liver, and evaluated their therapeutic effect in methionine-choline deficient diet (MCDD)-induced and high-fat-diet (HFD)-induced MASH model mice. Intravenously administered CO-RBCs effectively delivered CO to the MASH liver, where they prevented fat accumulation by promoting fatty acid oxidation via AMP-activated protein kinase (AMPK) activation and peroxisome proliferator-activated receptor induction. They also markedly suppressed Kupffer cell activation and their corresponding anti-inflammatory and antioxidative stress activities in MASH mice. CO-RBCs also helped to restore liver regeneration in mice with HFD-induced MASH by activating AMPK. We confirmed the underlying mechanisms by performing in vitro experiments in RAW264.7 cells and palmitate-stimulated HepG2 cells. Taken together, CO-RBCs show potential as a promising cellular treatment for MASH.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Monóxido de Carbono , Modelos Animales de Enfermedad , Eritrocitos , Macrófagos del Hígado , Enfermedad del Hígado Graso no Alcohólico , Animales , Macrófagos del Hígado/metabolismo , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Monóxido de Carbono/metabolismo , Humanos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Eritrocitos/metabolismo , Masculino , Hemo-Oxigenasa 1/metabolismo , Estrés Oxidativo/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Hígado/patología
15.
Nat Cardiovasc Res ; 3(3): 356-371, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-39196121

RESUMEN

Apolipoprotein-B (APOB)-containing lipoproteins cause atherosclerosis. Whether the vasculature is the initially responding site or if atherogenic dyslipidemia affects other organs simultaneously is unknown. Here we show that the liver responds to a dyslipidemic insult based on inducible models of familial hypercholesterolemia and APOB tracing. An acute transition to atherogenic APOB lipoprotein levels resulted in uptake by Kupffer cells and rapid accumulation of triglycerides and cholesterol in the liver. Bulk and single-cell RNA sequencing revealed a Kupffer-cell-specific transcriptional program that was not activated by a high-fat diet alone or detected in standard liver function or pathological assays, even in the presence of fulminant atherosclerosis. Depletion of Kupffer cells altered the dynamic of plasma and liver lipid concentrations, indicating that these liver macrophages help restrain and buffer atherogenic lipoproteins while simultaneously secreting atherosclerosis-modulating factors into plasma. Our results place Kupffer cells as key sentinels in organizing systemic responses to lipoproteins at the initiation of atherosclerosis.


Asunto(s)
Aterosclerosis , Modelos Animales de Enfermedad , Macrófagos del Hígado , Hígado , Macrófagos del Hígado/metabolismo , Animales , Hígado/metabolismo , Hígado/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Masculino , Hiperlipoproteinemia Tipo II/metabolismo , Hiperlipoproteinemia Tipo II/complicaciones , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/patología , Dislipidemias/metabolismo , Ratones Endogámicos C57BL , Triglicéridos/sangre , Triglicéridos/metabolismo , Apolipoproteínas B/metabolismo , Apolipoproteínas B/sangre , Colesterol/metabolismo , Colesterol/sangre , Dieta Alta en Grasa/efectos adversos , Apolipoproteína B-100/metabolismo , Femenino
16.
Chemosphere ; 364: 143032, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39111678

RESUMEN

Nano and microplastics are defined as particles smaller than 100 nm and 5 mm respectively. The widespread production and use of plastics in everyday life has resulted in significant accumulation of plastic debris in the environment. Over the last two decades there are increased concerns regarding the potential entry and accumulation of plastics in the human body with ingestion being one of the most important routes of exposure. However, the magnitude and nature of potential toxic effects of plastic exposure to human health is not yet fully understood. The liver is the body's principal detoxification organ and critically to this study recognized as the main accumulation site for particulates. In this study as the first of its kind the health impacts of long term low repeated polystyrene microplastics (1 and 5 µm) exposure was investigated in a functionally active 3D liver microtissue model, composed of primary human hepatocytes, Kupffer cells, sinusoidal endothelial cells and hepatic stellate cells. The highlight from the data includes microplastic-induced dose (3.125-25 µg/ml) and time dependent (up to 504 h) increase in cell death and inflammation manifested by enhanced release of IL6, IL8 and TNF-α. The exposure to repeated dosing of the plastics also resulted in notable pathology manifested as aberrant tissue architecture, such as dilated bile canaliculi and large lipid droplets inside the hepatic cells. This toxicity matched extremely well to the accumulation of the materials with the cells of microtissue predominately in the organ macrophages. This study highlights the real issue and danger of microplastic exposure with potential for long-term accumulation and adverse effects of non-biodegradable plastics within the liver.


Asunto(s)
Hepatocitos , Macrófagos del Hígado , Hígado , Microplásticos , Humanos , Microplásticos/toxicidad , Hígado/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Macrófagos del Hígado/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Poliestirenos/toxicidad , Células Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo
17.
Exp Mol Med ; 56(8): 1843-1855, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39122845

RESUMEN

Innate immune activation is critical for initiating hepatic inflammation during nonalcoholic steatohepatitis (NASH) progression. However, the mechanisms by which immunoregulatory molecules recognize lipogenic, fibrotic, and inflammatory signals remain unclear. Here, we show that high-fat diet (HFD)-induced oxidative stress activates Foxo1, YAP, and Notch1 signaling in hepatic macrophages. Macrophage Foxo1 deficiency (Foxo1M-KO) ameliorated hepatic inflammation, steatosis, and fibrosis, with reduced STING, TBK1, and NF-κB activation in HFD-challenged livers. However, Foxo1 and YAP double knockout (Foxo1/YAPM-DKO) or Foxo1 and Notch1 double knockout (Foxo1/Notch1M-DKO) promoted STING function and exacerbated HFD-induced liver injury. Interestingly, Foxo1M-KO strongly reduced TGF-ß1 release from palmitic acid (PA)- and oleic acid (OA)-stimulated Kupffer cells and decreased Col1α1, CCL2, and Timp1 expression but increased MMP1 expression in primary hepatic stellate cells (HSCs) after coculture with Kupffer cells. Notably, PA and OA challenge in Kupffer cells augmented LIMD1 and LATS1 colocalization and interaction, which induced YAP nuclear translocation. Foxo1M-KO activated PGC-1α and increased nuclear YAP activity, modulating mitochondrial biogenesis. Using chromatin immunoprecipitation (ChIP) coupled with massively parallel sequencing (ChIP-Seq) and in situ RNA hybridization, we found that NICD colocalizes with YAP and targets Mb21d1 (cGAS), while YAP functions as a novel coactivator of the NICD, which is crucial for reprogramming STING function in NASH progression. These findings highlight the importance of the macrophage Foxo1-YAP-Notch1 axis as a key molecular regulator that controls lipid metabolism, inflammation, and innate immunity in NASH.


Asunto(s)
Progresión de la Enfermedad , Proteína Forkhead Box O1 , Inmunidad Innata , Enfermedad del Hígado Graso no Alcohólico , Receptor Notch1 , Transducción de Señal , Proteínas Señalizadoras YAP , Animales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Proteína Forkhead Box O1/metabolismo , Receptor Notch1/metabolismo , Receptor Notch1/genética , Proteínas Señalizadoras YAP/metabolismo , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ratones Noqueados , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/inmunología , Dieta Alta en Grasa/efectos adversos , Macrófagos/metabolismo , Macrófagos/inmunología , Masculino , Modelos Animales de Enfermedad
18.
Cell Rep ; 43(8): 114629, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39146183

RESUMEN

In mice, the first liver-resident macrophages, known as Kupffer cells (KCs), are thought to derive from yolk sac (YS) hematopoietic progenitors that are specified prior to the emergence of the hematopoietic stem cell (HSC). To investigate human KC development, we recapitulated YS-like hematopoiesis from human pluripotent stem cells (hPSCs) and transplanted derivative macrophage progenitors into NSG mice previously humanized with hPSC-liver sinusoidal endothelial cells (LSECs). We demonstrate that hPSC-LSECs facilitate stable hPSC-YS-macrophage engraftment for at least 7 weeks. Single-cell RNA sequencing (scRNA-seq) of engrafted YS-macrophages revealed a homogeneous MARCO-expressing KC gene signature and low expression of monocyte-like macrophage genes. In contrast, human cord blood (CB)-derived macrophage progenitors generated grafts that contain multiple hematopoietic lineages in addition to KCs. Functional analyses showed that the engrafted KCs actively perform phagocytosis and erythrophagocytosis in vivo. Taken together, these findings demonstrate that it is possible to generate human KCs from hPSC-derived, YS-like progenitors.


Asunto(s)
Diferenciación Celular , Células Endoteliales , Macrófagos del Hígado , Hígado , Células Madre Pluripotentes , Humanos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Células Endoteliales/metabolismo , Células Endoteliales/citología , Animales , Hígado/citología , Hígado/metabolismo , Ratones , Fagocitosis , Hematopoyesis
19.
Proc Natl Acad Sci U S A ; 121(35): e2405746121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39172787

RESUMEN

While macrophage heterogeneity during metabolic dysfunction-associated steatohepatitis (MASH) has been described, the fate of these macrophages during MASH regression is poorly understood. Comparing macrophage heterogeneity during MASH progression vs regression, we identified specific macrophage subpopulations that are critical for MASH/fibrosis resolution. We elucidated the restorative pathways and gene signatures that define regression-associated macrophages and establish the importance of TREM2+ macrophages during MASH regression. Liver-resident Kupffer cells are lost during MASH and are replaced by four distinct monocyte-derived macrophage subpopulations. Trem2 is expressed in two macrophage subpopulations: i) monocyte-derived macrophages occupying the Kupffer cell niche (MoKC) and ii) lipid-associated macrophages (LAM). In regression livers, no new transcriptionally distinct macrophage subpopulation emerged. However, the relative macrophage composition changed during regression compared to MASH. While MoKC was the major macrophage subpopulation during MASH, they decreased during regression. LAM was the dominant macrophage subtype during MASH regression and maintained Trem2 expression. Both MoKC and LAM were enriched in disease-resolving pathways. Absence of TREM2 restricted the emergence of LAMs and formation of hepatic crown-like structures. TREM2+ macrophages are functionally important not only for restricting MASH-fibrosis progression but also for effective regression of inflammation and fibrosis. TREM2+ macrophages are superior collagen degraders. Lack of TREM2+ macrophages also prevented elimination of hepatic steatosis and inactivation of HSC during regression, indicating their significance in metabolic coordination with other cell types in the liver. TREM2 imparts this protective effect through multifactorial mechanisms, including improved phagocytosis, lipid handling, and collagen degradation.


Asunto(s)
Macrófagos del Hígado , Cirrosis Hepática , Macrófagos , Glicoproteínas de Membrana , Receptores Inmunológicos , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Animales , Ratones , Macrófagos/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Hígado/patología , Metabolismo de los Lípidos , Ratones Endogámicos C57BL , Masculino , Lípidos , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/genética , Ratones Noqueados
20.
Hepatol Commun ; 8(9)2024 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-39185911

RESUMEN

BACKGROUND: Dysregulated fatty acid metabolism is closely linked to the development of alcohol-associated liver disease (ALD). KCs, which are resident macrophages in the liver, play a critical role in ALD pathogenesis. However, the effect of alcohol on fatty acid metabolism in KCs remains poorly understood. The current study aims to investigate fatty acid metabolism in KCs and its potential effect on ALD development. METHODS: Wild-type C57BL/6 mice were fed a Lieber-DeCarli ethanol liquid diet for 3 days. Then, the liver injury and levels of intrahepatic bacteria were assessed. Next, we investigated the effects and underlying mechanisms of ethanol exposure on fatty acid metabolism and the phagocytosis of KCs, both in vivo and in vitro. Finally, we generated KCs-specific Fasn knockout and overexpression mice to evaluate the impact of FASN on the phagocytosis of KCs and ethanol-induced liver injury. RESULTS: Using Bodipy493/503 to stain intracellular neutral lipids, we found significantly reduced lipid levels in KCs from mice fed an alcohol-containing diet for 3 days and in RAW264.7 macrophages exposed to ethanol. Mechanistically, alcohol exposure suppressed sterol regulatory element-binding protein 1 transcriptional activity, thereby inhibiting fatty acid synthase (FASN)-mediated de novo lipogenesis in macrophages both in vitro and in vivo. We show that genetic ablation and pharmacologic inhibition of FASN significantly impaired KC's ability to take up and eliminate bacteria. Conversely, KCs-specific Fasn overexpression reverses the impairment of macrophage phagocytosis caused by alcohol exposure. We also revealed that KCs-specific Fasn knockout augmented KCs apoptosis and exacerbated liver injury in mice fed an alcohol-containing diet for 3 days. CONCLUSIONS: Our findings indicate the crucial role of de novo lipogenesis in maintaining effective KCs phagocytosis and suggest a therapeutic target for ALD based on fatty acid synthesis in KCs.


Asunto(s)
Ácidos Grasos , Macrófagos del Hígado , Hepatopatías Alcohólicas , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Animales , Macrófagos del Hígado/metabolismo , Ratones , Ácidos Grasos/metabolismo , Ácidos Grasos/biosíntesis , Hepatopatías Alcohólicas/metabolismo , Etanol , Acido Graso Sintasa Tipo I/metabolismo , Acido Graso Sintasa Tipo I/genética , Masculino , Progresión de la Enfermedad , Hígado/metabolismo , Lipogénesis/efectos de los fármacos , Células RAW 264.7 , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...