Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 409
Filtrar
1.
Hepatol Commun ; 8(9)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39225682

RESUMEN

Hepatocellular carcinoma (HCC), the major type of primary liver cancer, is notorious for its resistance to systemic treatments. The field has made a great leap in the past decade, with the number of FDA-approved therapies for advanced HCC increasing from 1 to 9. Although tyrosine kinase inhibitors remain the most common first-line option as monotherapy treatment, the clinical success of immune checkpoint inhibitors, especially when used in combination with anti-VEGF/VEGFR in HCC will likely transform the treatment landscape. While immune checkpoint inhibitors represent an exciting therapeutic revenue for HCC, recent studies have revealed that nonviral HCC, which is primarily caused by metabolic dysfunction-associated steatotic hepatitis (MASH), has a distinct and less favorable response to the immune checkpoint inhibitors. MASH is the most rapidly increasing etiology for HCC. The immune microenvironment of MASH-HCC is greatly affected by the intertwined pathological processes of steatosis-induced iterative cycles between steatohepatitis and liver injury. Here, we present a timely summary of the immune microenvironment of MASH-HCC. We will delve into the use of cutting-edge technologies, such as single-cell RNA sequencing, spatial transcriptomics, and mass cytometry imaging, to deconvolute the complexity of the immune ecosystem in MASH-HCC. We will also discuss the novel therapeutic innovations for MASH-HCC in preclinical models, such as the metabolic inhibitor, epigenetic inhibitor, and immunomodulator. These inhibitors all have the ability to subvert the immune microenvironment of MASH-HCC, improving the efficiency of anti-PD-1. While awaiting new drugs to be tested in clinical trials, the knowledge gained from these investigations is crucial for the development of personalized and effective treatment strategies for MASH-HCC.


Asunto(s)
Carcinoma Hepatocelular , Inhibidores de Puntos de Control Inmunológico , Neoplasias Hepáticas , Microambiente Tumoral , Humanos , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/tratamiento farmacológico , Microambiente Tumoral/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Hígado Graso/inmunología
2.
Lipids Health Dis ; 23(1): 249, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39148061

RESUMEN

BACKGROUND: Existing studies have presented limited and disparate findings on the nexus between immune cells, plasma metabolites, and metabolic dysfunction-associated steatotic liver disease (MASLD). The aim of this study was to investigate the causal relationship between immune cells and MASLD. Additionally, we aimed to identify and quantify the potential mediating role of metabolites. METHODS: A Mendelian randomization (MR) analysis was conducted using two samples of pooled data from genome-wide association studies on MASLD that included 2568 patients and 409,613 control individuals. Additionally, a mediated MR study was employed to quantify the metabolite-mediated immune cell effects on MASLD. RESULTS: In this study, eight immunophenotypes were linked to the risk of MASLD, and thirty-five metabolites/metabolite ratios were linked to the occurrence of MASLD. Furthermore, a total of six combinations of immunophenotypic and metabolic factors demonstrated effects on the occurrence of MASLD, although the mediating effects of metabolites were not significant. CONCLUSION: Our study demonstrated that certain immunophenotypes and metabolite/metabolite ratios have independent causal relationships with MASLD. Furthermore, we identified specific metabolites/metabolite ratios that are associated with an increased risk of MASLD. However, their mediating role in the causal association between immunophenotypes and MASLD was not significant. It is important to consider immune and metabolic disorders among patients with MASLD in clinical practice.


Asunto(s)
Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Humanos , Polimorfismo de Nucleótido Simple , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/inmunología , Inmunofenotipificación , Masculino
3.
Int J Mol Sci ; 25(15)2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39125804

RESUMEN

Obesity is an emerging public health problem. Chronic low-grade inflammation is considered a major promotor of obesity-induced secondary diseases such as cardiovascular and fatty liver disease, type 2 diabetes mellitus, and several cancer entities. Most preliminary studies on obesity-induced immune responses have been conducted in male rodents. Sex-specific differences between men and women in obesity-induced immune dysregulation have not yet been fully outlined but are highly relevant to optimizing prevention strategies for overweight-associated complications. In this study, we fed C57BL/6 female vs. male mice with either standard chow or an obesity-inducing diet (OD). Blood and spleen immune cells were isolated and analyzed by flow cytometry. Lean control mice showed no sex bias in systemic and splenic immune cell composition, whereas the immune responses to obesity were significantly distinct between female and male mice. While immune cell alterations in male OD mice were characterized by a significant reduction in T cells and an increase in myeloid-derived suppressor cells (MDSC), female OD mice displayed preserved T cell numbers. The sex-dependent differences in obesity-induced T cell dysregulation were associated with varying susceptibility to body weight gain and fatty liver disease: Male mice showed significantly more hepatic inflammation and histopathological stigmata of fatty liver in comparison to female OD mice. Our findings indicate that sex impacts susceptibility to obesity-induced T cell dysregulation, which might explain sex-dependent different incidences in the development of obesity-associated secondary diseases. These results provide novel insights into the understanding of obesity-induced chronic inflammation from a sex-specific perspective. Given that most nutrition, exercise, and therapeutic recommendations for the prevention of obesity-associated comorbidities do not differentiate between men and women, the data of this study are clinically relevant and should be taken into consideration in future trials and treatment strategies.


Asunto(s)
Ratones Endogámicos C57BL , Obesidad , Linfocitos T , Animales , Obesidad/inmunología , Obesidad/complicaciones , Obesidad/etiología , Femenino , Masculino , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Dieta Alta en Grasa/efectos adversos , Factores Sexuales , Bazo/inmunología , Bazo/patología , Caracteres Sexuales , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Hígado Graso/etiología , Hígado Graso/inmunología , Hígado Graso/patología , Inflamación/inmunología , Inflamación/patología , Inflamación/etiología
4.
Front Immunol ; 15: 1437046, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39156888

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.


Asunto(s)
Interleucina-17 , Interleucina-22 , Interleucinas , Humanos , Interleucina-17/metabolismo , Interleucina-17/inmunología , Interleucinas/metabolismo , Interleucinas/inmunología , Animales , Hígado Graso/inmunología , Hígado Graso/metabolismo , Hígado Graso/patología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/inmunología , Hígado/patología , Hígado/metabolismo , Hígado/inmunología
5.
J Pediatr Gastroenterol Nutr ; 79(3): 667-673, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38966965

RESUMEN

Significance of autoantibodies in pediatric metabolic dysfunction-associated steatotic liver disease (MASLD) is unknown. Our aim was to determine the prevalence and significance of autoantibodies in MASLD. PubMed and Scopus were searched and six articles (689 [487 males] MASLD patients) were identified. Antinuclear antibodies (ANA) was positive in 28% (95% confidence interval [CI]: 17%-39%, n = 6 studies), antismooth muscle antibodies (ASMA) in 28% (95% CI: 8%-50%, n = 5 studies), actin-positive in 15% (95% CI: 10%-20%, n = 2 studies) and elevated immunoglobulin G in 17% (95% CI: 1%-39%, n = 4 studies). Anti-liver-kidney-microsomal antibody was not present in any patient. There was no significant association of ANA positivity with degree of liver steatosis, liver fibrosis or nonalcoholic fatty liver disease activity score (NAS) but patients with ASMA positivity had advanced fibrosis (pooled risk ratio [RR] 1.77; 95% CI 1.16-2.71) and higher risk of NAS ≥5 (pooled RR 1.21; 95% CI: 1.01-1.44, n = 2 studies, 243 patients). To conclude, non-organ specific autoantibodies are present in over one-fourth of children with MASLD and the presence of ASMA maybe associated with increased disease severity.


Asunto(s)
Autoanticuerpos , Humanos , Autoanticuerpos/sangre , Niño , Prevalencia , Hígado Graso/inmunología , Hígado Graso/epidemiología , Masculino , Femenino
6.
J Immunol ; 213(3): 251-256, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39008791

RESUMEN

Metabolic dysfunction-associated steatohepatitis (MASH) is characterized by infiltration of monocyte-derived macrophages (MdMs) into the liver; however, the function of these macrophages is largely unknown. We previously demonstrated that a population of MdMs, referred to as hepatic lipid-associated macrophages (LAMs), assemble into aggregates termed hepatic crown-like structures in areas of liver fibrosis. Intriguingly, decreasing MdM recruitment resulted in increased liver fibrosis, suggesting that LAMs contribute to antifibrotic pathways in MASH. In this study, we determined that hepatic crown-like structures are characterized by intimate interactions between activated hepatic stellate cells (HSCs) and macrophages in a collagen matrix in a mouse model of MASH. MASH macrophages displayed collagen-degrading capacities, and HSCs derived from MASH livers promoted expression of LAM marker genes and acquisition of a collagen-degrading phenotype in naive macrophages. These data suggest that crosstalk between HSCs and macrophages may contribute to collagen degradation MASH.


Asunto(s)
Células Estrelladas Hepáticas , Cirrosis Hepática , Macrófagos , Fenotipo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/inmunología , Células Estrelladas Hepáticas/patología , Animales , Ratones , Macrófagos/inmunología , Macrófagos/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/inmunología , Ratones Endogámicos C57BL , Colágeno/metabolismo , Modelos Animales de Enfermedad , Humanos , Hígado/patología , Hígado/metabolismo , Hígado/inmunología , Masculino , Hígado Graso/patología , Hígado Graso/metabolismo , Hígado Graso/inmunología
7.
Nutrients ; 16(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892602

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent type of liver disease worldwide. The exact pathophysiology behind MASLD remains unclear; however, it is thought that a combination of factors or "hits" act as precipitants for disease onset and progression. Abundant evidence supports the roles of diet, genes, metabolic dysregulation, and the intestinal microbiome in influencing the accumulation of lipids in hepatocytes and subsequent progression to inflammation and fibrosis. Currently, there is no cure for MASLD, but lifestyle changes have been the prevailing cornerstones of management. Research is now focusing on the intestinal microbiome as a potential therapeutic target for MASLD, with the spotlight shifting to probiotics, antibiotics, and fecal microbiota transplantation. In this review, we provide an overview of how intestinal microbiota interact with the immune system to contribute to the pathogenesis of MASLD and metabolic dysfunction-associated steatohepatitis (MASH). We also summarize key microbial taxa implicated in the disease and discuss evidence supporting microbial-targeted therapies in its management.


Asunto(s)
Progresión de la Enfermedad , Microbioma Gastrointestinal , Humanos , Trasplante de Microbiota Fecal , Sistema Inmunológico/metabolismo , Probióticos/uso terapéutico , Hígado Graso/microbiología , Hígado Graso/inmunología , Animales , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Antibacterianos/uso terapéutico , Hígado/metabolismo
8.
Trends Pharmacol Sci ; 45(7): 614-627, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38853100

RESUMEN

Growing evidence suggests that metabolic dysfunction-associated steatotic liver disease (MASLD) is significantly higher in men versus women. Increased prevalence is observed in postmenopausal women, suggesting that age and sex (hormones) influence MASLD development and progression. Molecular data further reveal that sex regulates the innate immune responses with an essential role in MASLD progression. To date, there has been limited focus on the role of innate immune sexual dimorphism in MASLD, and differences between men and women are not considered in the current drug discovery landscape. In this review, we summarize the sex disparities and innate immune sexual dimorphism in MASLD pathogenesis. We further highlight the importance of harnessing sexual dimorphism in identifying therapeutic targets, developing pharmacological therapies, and designing (pre-) clinical studies for the personalized treatment for MASLD.


Asunto(s)
Inmunidad Innata , Caracteres Sexuales , Humanos , Animales , Femenino , Masculino , Hígado Graso/inmunología , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/tratamiento farmacológico
9.
Cell Rep Med ; 5(5): 101564, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38733988

RESUMEN

Monocytes (Mos) are crucial in the evolution of metabolic dysfunction-associated steatotic liver disease (MASLD) to metabolic dysfunction-associated steatohepatitis (MASH), and immunometabolism studies have recently suggested targeting leukocyte bioenergetics in inflammatory diseases. Here, we reveal a peculiar bioenergetic phenotype in circulating Mos of patients with MASH, characterized by high levels of glycolysis and mitochondrial (mt) respiration. The enhancement of mt respiratory chain activity, especially complex II (succinate dehydrogenase [SDH]), is unbalanced toward the production of reactive oxygen species (ROS) and is sustained at the transcriptional level with the involvement of the AMPK-mTOR-PGC-1α axis. The modulation of mt activity with dimethyl malonate (DMM), an SDH inhibitor, restores the metabolic profile and almost abrogates cytokine production. Analysis of a public single-cell RNA sequencing (scRNA-seq) dataset confirms that in murine models of MASH, liver Mo-derived macrophages exhibit an upregulation of mt and glycolytic energy pathways. Accordingly, the DMM injection in MASH mice contrasts Mo infiltration and macrophagic enrichment, suggesting immunometabolism as a potential target in MASH.


Asunto(s)
Metabolismo Energético , Mitocondrias , Monocitos , Humanos , Animales , Monocitos/metabolismo , Monocitos/inmunología , Ratones , Mitocondrias/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/inmunología , Masculino , Glucólisis , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Macrófagos/inmunología , Femenino , Hígado/metabolismo , Hígado/patología
10.
Biomolecules ; 14(4)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38672492

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most common liver disease worldwide in recent years. MASLD commonly presents as simple hepatic steatosis, but ~25% of patients develop liver inflammation, progressive fibrosis, liver cirrhosis and related hepatocellular carcinoma. Liver inflammation and the degree of fibrosis are key determinants of the prognosis. The pathophysiology of liver inflammation is incompletely understood and involves diverse factors and specifically innate and adaptive immune responses. More specifically, diverse mediators of innate immunity such as proinflammatory cytokines, adipokines, inflammasomes and various cell types like mononuclear cells, macrophages and natural killer cells are involved in directing the inflammatory process in MASLD. The activation of innate immunity is driven by various factors including excess lipids and lipotoxicity, insulin resistance and molecular patterns derived from gut commensals. Targeting pathways of innate immunity might therefore appear as an attractive therapeutic strategy in the future management of MASLD and possibly its complications.


Asunto(s)
Inmunidad Innata , Humanos , Animales , Hígado Graso/inmunología , Inflamasomas/inmunología , Inflamasomas/metabolismo , Citocinas/metabolismo , Citocinas/inmunología , Resistencia a la Insulina/inmunología , Inflamación/inmunología
11.
Front Immunol ; 15: 1379225, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38650949

RESUMEN

Dendritic cells (DCs) are major antigen-presenting cells that connect innate and adaptive immunity. Hepatic DCs are less activated and contribute to maintain the tolerogenic environment of the liver under steady state. Several studies indicated DCs in metabolic dysfunction-associated steatohepatitis (MASH), representing a substantial burden on healthcare systems due to its association with liver-related morbidity and mortality. Studies highlighted the potential disease-promoting role of liver DCs in the development of MASH while other experimental systems suggested their protective role. This review discusses this controversy and the current understanding of how DCs affect the pathogenesis of MASH.


Asunto(s)
Células Dendríticas , Células Dendríticas/inmunología , Humanos , Animales , Hígado/inmunología , Hígado Graso/inmunología
12.
Int Immunopharmacol ; 133: 112091, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38657500

RESUMEN

The pathophysiology of hepatic steatosis is thoroughly reviewed in this comprehensive report, with particular attention to the complex interactions between inflammatory pathways, insulin resistance, lipid metabolism, metabolic dysregulation, and immunological responses in the liver including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC). The study highlights the role of immune cell regulation in disease progression and explores the potential of immune cell-specific treatments for treating hepatic disorders. The development of liver disorders is significantly influenced by immune cells, including dendritic cells, T cells, and natural killer cells. Clinical investigations show that immune cell-specific treatments can effectively reduce liver fibrosis and inflammation. Future research should focus on finding new immunological targets for therapeutic interventions, as well as addressing the management challenges associated with NAFLD/NASH. Hepatic immune microorganisms also impact liver homeostasis and disorders. Improvements in immune cell regulation and liver transplantation methods give patients hope for better prognoses. Important phases include optimizing the selection of donors for malignancy of the liver, using machine perfusion for organ preservation, and fine-tuning immunosuppressive strategies. For focused treatments in hepatic steatosis, it is imperative to understand the intricate interactions between immune and metabolic variables. Understanding the liver's heterogeneous immune profile, encompassing a range of immune cell subpopulations, is crucial for formulating focused therapeutic interventions. To improve patient care and outcomes in hepatic illnesses, there is an urgent need for further research and innovation. Therefore, to effectively treat hepatic steatosis, it is important to enhance therapeutic techniques and maximize liver transplantation strategies.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Animales , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/terapia , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/terapia , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Metabolismo de los Lípidos , Resistencia a la Insulina/inmunología , Hígado Graso/inmunología , Hígado Graso/metabolismo , Hígado Graso/terapia
13.
J Nutr Biochem ; 99: 108852, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34525389

RESUMEN

This study aimed to develop a well-characterized mouse model of alcoholic hepatitis (AH) regression. Male C57BL/6J mice were fed a Lieber-DeCarli (LD) control diet or LD containing 5% ethanol for ten days followed by one binge, which is the chronic-binge model of AH developed by the National Institute on Alcohol Abuse and Alcoholism. To determine AH regression, mice previously exposed to ethanol were put on LD control diet and metabolic and inflammatory features were monitored weekly for three weeks. Serum alcohol, total cholesterol, and alanine transaminase levels were increased in ethanol-fed mice, which declined to those of no ethanol controls within one and three weeks after ethanol withdrawal, respectively. Serum malondialdehyde was increased with ethanol feeding, but it was restored to no ethanol control levels within one week. Ethanol-induced changes in the hepatic expression of genes involved in lipogenesis, fatty acid oxidation, ethanol metabolism, and antioxidant response were restored to those of no ethanol controls after 3 weeks of ethanol withdrawal. Also, ethanol-induced hepatic inflammation was gradually decreased during the 3 weeks of ethanol withdrawal. Hepatic nicotinamide adenine dinucleotide (NAD+) levels and the expression of enzymes involved in the NAD+ salvage pathway were decreased by ethanol feeding, which was mitigated after ethanol withdrawal. Ethanol significantly lowered hepatic sirtuin 1 expression, but its levels were restored with ethanol cessation. This study established a mouse model of AH regression, which can be used as a preclinical model to study the potential of dietary bioactives or therapeutic agents on AH regression.


Asunto(s)
Alcoholismo/complicaciones , Etanol/efectos adversos , Hígado Graso/metabolismo , Hepatitis Alcohólica/metabolismo , NAD/metabolismo , Estrés Oxidativo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/inmunología , Hepatitis Alcohólica/etiología , Hepatitis Alcohólica/genética , Hepatitis Alcohólica/inmunología , Humanos , Hígado/inmunología , Hígado/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Sirtuina 1/genética , Sirtuina 1/metabolismo
14.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884762

RESUMEN

Immune checkpoint inhibitors represent one of the most significant recent advances in clinical oncology, since they dramatically improved the prognosis of deadly cancers such as melanomas and lung cancer. Treatment with these drugs may be complicated by the occurrence of clinically-relevant adverse drug reactions, most of which are immune-mediated, such as pneumonitis, colitis, endocrinopathies, nephritis, Stevens Johnson syndrome and toxic epidermal necrolysis. Drug-induced steatosis and steatohepatitis are not included among the typical forms of cancer immunotherapy-induced liver toxicity, which, instead, usually occurs as a panlobular hepatitis with prominent lymphocytic infiltrates. Nonetheless, non-alcoholic fatty liver disease is a risk factor for immunotherapy-induced hepatitis, and steatosis and steatohepatitis are frequently observed in this condition. In the present review we discuss how these pathology findings could be explained in the context of current models suggesting immune-mediated pathogenesis for steatohepatitis. We also review evidence suggesting that in patients with hepatocellular carcinoma, the presence of steatosis or steatohepatitis could predict a poor therapeutic response to these agents. How these findings could fit with immune-mediated mechanisms of these liver diseases will also be discussed.


Asunto(s)
Hígado Graso/etiología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia/efectos adversos , Neoplasias/terapia , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/terapia , Hígado Graso/inmunología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Modelos Biológicos , Neoplasias/inmunología , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Pronóstico , Factores de Riesgo
15.
Front Immunol ; 12: 726829, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691032

RESUMEN

ß2 integrins are critical for neutrophil firm adhesion, trans-endothelial migration, and the recruitment to the inflamed tissue. Autophagy is implicated in cell migration and tumor metastasis through facilitating the turnover of ß1 integrins; however, whether autophagy is able to control neutrophil migration by promoting the degradation of ß2 integrins is unexplored. Here, we show that high blood levels of palmitic acid (PA) strongly triggered neutrophil autophagy activation, leading to adhesion deficiency in dairy cows with fatty liver. The three neutrophil granule subtypes, namely, azurophil granules (AGs), specific granules (SGs), and gelatinase granules (GGs), were engulfed by the autophagosomes for degradation, resulting in an increased vacuolation in fatty liver dairy cow neutrophils. Importantly, the adhesion-associated molecules CD11b and CD18 distributed on AGs, SGs, and GGs were degraded with the three granule subtypes by autophagy. Moreover, FGA, Hsc70, and TRIM21 mediated the degradation of cytosolic oxidized-ubiquitinated CD11b and CD18. Collectively, our results demonstrate that high blood PA triggers neutrophil autophagy-dependent vacuolation and granule-dependent adhesion deficiency, decreasing neutrophil mobility, and impairing the innate immune system of dairy cow with fatty liver. This theory extends the category of autophagy in maintaining granule homeostasis and provides a novel strategy to improve the immune of dairy cows with metabolic disease.


Asunto(s)
Autofagia , Adhesión Celular , Hígado Graso/inmunología , Neutrófilos/fisiología , Ácido Palmítico/sangre , Animales , Proteína 5 Relacionada con la Autofagia/genética , Antígeno CD11b/inmunología , Antígenos CD18/inmunología , Bovinos , Hígado Graso/sangre , Femenino , Fibrinógeno/genética , Células HL-60 , Proteínas del Choque Térmico HSC70/genética , Humanos , Antígeno de Macrófago-1 , Ribonucleoproteínas/genética
16.
Cell Prolif ; 54(10): e13116, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34469018

RESUMEN

OBJECTIVES: The discrepancy between supply and demand of organ has led to an increased utilization of steatotic liver for liver transplantation (LT). Hepatic steatosis, however, is a major risk factor for graft failure due to increased susceptibility to ischaemia-reperfusion (I/R) injury during transplantation. MATERIALS AND METHODS: To assess the plasticity and phenotype of immune cells within the microenvironment of steatotic liver graft at single-cell level, single-cell RNA-sequencing (scRNA-Seq) was carried out on 23 675 cells from transplanted rat livers. Bioinformatic analyses and multiplex immunohistochemistry were performed to assess the functional properties, transcriptional regulation, phenotypic switching and cell-cell interactions of different cell subtypes. RESULTS: We have identified 11 different cell types in transplanted livers and found that the highly complex ecosystem was shaped by myeloid-derived cell subsets that transit between different states and interact mutually. Notably, a pro-inflammatory phenotype of Kupffer cells (KCs) with high expression of colony-stimulating factor 3 (CSF3) that was enriched in transplanted steatotic livers was potentially participated in fatty graft injury. We have also detected a subset of dendritic cells (DCs) with highly expressing XCR1 that was correlated with CD8+ T cells, mediating the severer steatotic liver damage by I/R injury. CONCLUSIONS: The findings of our study provide new insight into the mechanisms by which steatosis exacerbates liver damage from I/R injury. Interventions based on these observations create opportunities in attenuating fatty liver graft injury and expanding the donor pool.


Asunto(s)
Hígado Graso/inmunología , Hígado/inmunología , Daño por Reperfusión/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Macrófagos del Hígado/inmunología , Trasplante de Hígado/métodos , Fenotipo , Ratas , Ratas Sprague-Dawley , Análisis de la Célula Individual/métodos , Transcripción Genética/inmunología
17.
Front Immunol ; 12: 694720, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248992

RESUMEN

The present study aimed to elucidate the mechanism of dietary betaine, as a lipid-lowering substance, on the regulation of lipid metabolism and inflammation in juvenile black seabream (Acanthopagrus schlegelii) fed a high fat diet. An 8-week feeding trial was conducted in black seabream with an initial weight of 8.39 ± 0.01g fed four isonitrogenous diets including Control, medium-fat diet (11%); HFD, high-fat diet (17%); and HFD supplemented with two levels (10 and 20 g/kg) of betaine, HFD+B1 and HFD+B2, respectively. SGR and FE in fish fed HFD+B2 were significantly higher than in fish fed HFD. Liver histology revealed that vacuolar fat droplets were smaller and fewer in bream fed HFD supplemented with betaine compared to fish fed HFD. Betaine promoted the mRNA and protein expression levels of silent information regulator 1 (Sirt1), up-regulated mRNA expression and protein content of lipid peroxisome proliferator-activated receptor alpha (pparα), and down-regulated mRNA expression and protein content of sterol regulatory element-binding protein-1(srebp-1). Furthermore, the mRNA expression levels of anti-inflammatory cytokines in liver and intestine were up-regulated, while nuclear factor kB (nf-kb) and pro-inflammatory cytokines were down-regulated by dietary betaine supplementation. Likewise, in fish that received lipopolysaccharide (LPS) to stimulate inflammatory responses, the expression levels of mRNAs of anti-inflammatory cytokines in liver, intestine and kidney were up-regulated in fish fed HFD supplemented with betaine compared with fish fed HFD, while nf-kb and pro-inflammatory cytokines were down-regulated. This is the first report to suggest that dietary betaine could be an effective feed additive to alleviate hepatic steatosis and attenuate inflammatory responses in black seabream fed a high fat diet by modulating the Sirt1/Srebp-1/Pparɑ pathway.


Asunto(s)
Betaína/administración & dosificación , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Hígado Graso/veterinaria , Enfermedades de los Peces/prevención & control , Proteínas de Peces/metabolismo , Inflamación/veterinaria , Hígado/enzimología , PPAR alfa/metabolismo , Dorada/metabolismo , Sirtuina 1/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factores de Edad , Alimentación Animal , Animales , Citocinas/genética , Citocinas/metabolismo , Hígado Graso/enzimología , Hígado Graso/inmunología , Hígado Graso/prevención & control , Enfermedades de los Peces/enzimología , Enfermedades de los Peces/inmunología , Proteínas de Peces/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/prevención & control , Hígado/inmunología , PPAR alfa/genética , Dorada/genética , Dorada/inmunología , Sirtuina 1/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
18.
Sci Rep ; 11(1): 12314, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112902

RESUMEN

We tested the hypothesis that a particular immune activation profile might be correlated with insulin resistance in a general population. By measuring 43 markers of immune, endothelial, and coagulation activation, we have previously shown that five different immune activation profiles may be distinguished in 150 volunteers. One of these profiles, Profile 2, characterized by CD4+ T cell senescence, inflammation, monocyte, B cell, and endothelial activation, presented elevated insulinemia, glycemia, triglyceridemia, and γ-glutamyl transferase, a marker of liver injury, in comparison with other profiles. Our data are compatible with a model in which a particular immune activation profile might favor the development of insulin resistance and metabolic syndrome. In this hypothesis, identification of this profile, that is feasible with only 3 markers with an error rate of 5%, might allow to personalize the screening and prevention of metabolic syndrome-driven morbidities as liver steatosis.


Asunto(s)
Inflamación/inmunología , Resistencia a la Insulina/inmunología , Síndrome Metabólico/inmunología , Linfocitos T/inmunología , gamma-Glutamiltransferasa/genética , Anciano , Linfocitos B/inmunología , Biomarcadores/sangre , Glucemia , Linfocitos T CD4-Positivos/inmunología , Senescencia Celular/genética , Hígado Graso/genética , Hígado Graso/inmunología , Femenino , Humanos , Inflamación/genética , Inflamación/patología , Resistencia a la Insulina/genética , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Persona de Mediana Edad , Monocitos/inmunología , Linfocitos T/patología
19.
Nat Med ; 27(6): 1043-1054, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34017133

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are prevalent liver conditions that underlie the development of life-threatening cirrhosis, liver failure and liver cancer. Chronic necro-inflammation is a critical factor in development of NASH, yet the cellular and molecular mechanisms of immune dysregulation in this disease are poorly understood. Here, using single-cell transcriptomic analysis, we comprehensively profiled the immune composition of the mouse liver during NASH. We identified a significant pathology-associated increase in hepatic conventional dendritic cells (cDCs) and further defined their source as NASH-induced boost in cycling of cDC progenitors in the bone marrow. Analysis of blood and liver from patients on the NAFLD/NASH spectrum showed that type 1 cDCs (cDC1) were more abundant and activated in disease. Sequencing of physically interacting cDC-T cell pairs from liver-draining lymph nodes revealed that cDCs in NASH promote inflammatory T cell reprogramming, previously associated with NASH worsening. Finally, depletion of cDC1 in XCR1DTA mice or using anti-XCL1-blocking antibody attenuated liver pathology in NASH mouse models. Overall, our study provides a comprehensive characterization of cDC biology in NASH and identifies XCR1+ cDC1 as an important driver of liver pathology.


Asunto(s)
Células Dendríticas/inmunología , Hígado Graso/inmunología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Receptores de Quimiocina/genética , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Células Dendríticas/patología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/patología , Femenino , Humanos , Hígado/inmunología , Hígado/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Receptores de Quimiocina/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
20.
Front Immunol ; 12: 651728, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859644

RESUMEN

The coronavirus infectious disease 2019 (COVID-19) pandemic has hit the world, affecting health, medical care, economies and our society as a whole. Furthermore, COVID-19 pandemic joins the increasing prevalence of metabolic syndrome in western countries. Patients suffering from obesity, type II diabetes mellitus, cardiac involvement and metabolic associated fatty liver disease (MAFLD) have enhanced risk of suffering severe COVID-19 and mortality. Importantly, up to 25% of the population in western countries is susceptible of suffering from both MAFLD and COVID-19, while none approved treatment is currently available for any of them. Moreover, it is well known that exacerbated innate immune responses are key in the development of the most severe stages of MAFLD and COVID-19. In this review, we focus on the role of the immune system in the establishment and progression of MAFLD and discuss its potential implication in the development of severe COVID-19 in MAFLD patients. As a result, we hope to clarify their common pathology, but also uncover new potential therapeutic targets and prognostic biomarkers for further research.


Asunto(s)
Inmunidad Adaptativa/inmunología , COVID-19/inmunología , COVID-19/patología , Hígado Graso/inmunología , Inmunidad Innata/inmunología , Enzima Convertidora de Angiotensina 2/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 2/patología , Hígado Graso/patología , Humanos , Hígado/inmunología , Hígado/patología , Obesidad/patología , Factores de Riesgo , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA