Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 562
Filtrar
1.
Hepatol Commun ; 8(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38967563

RESUMEN

The liver is a vital organ that continuously adapts to a wide and dynamic diversity of self-antigens and xenobiotics. This involves the active contribution of immune cells, particularly by the liver-resident macrophages, the Kupffer cells (KCs), which exert a variety of central functions in liver homeostasis and disease. As such, KCs interact with their microenvironment to shape the hepatic cellular landscape, control gut-derived signal integration, and modulate metabolism. On injury, the rapid recruitment of bone marrow monocyte-derived macrophages alters this status quo and, when unrestrained, drastically compromises liver homeostasis, immune surveillance, and tissue organization. Several factors determine the functional roles of liver macrophages in these processes, such as their ontogeny, activation/polarization profile and, importantly, spatial distribution within the liver. Loss of tolerance and adaptability of the hepatic immune environment may result in persistent inflammation, hepatic fibrosis, cirrhosis, and a tumorigenic niche promoting liver cancer. In this review, we aim at providing the most recent breakthroughs in our understanding of liver macrophage biology, particularly their diversity and adaptability in the hepatic spatiotemporal context, as well as on potential therapeutic interventions that may hold the key to tackling remaining clinical challenges of varying etiologies in hepatology.


Asunto(s)
Macrófagos del Hígado , Hígado , Humanos , Hígado/inmunología , Hígado/patología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/fisiología , Animales , Macrófagos/inmunología , Macrófagos/fisiología , Homeostasis/inmunología
2.
Stem Cell Res Ther ; 15(1): 48, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378583

RESUMEN

BACKGROUND: Allogeneic hepatocyte transplantation is an emerging approach to treat acute liver defects. However, durable engraftment of the transplanted cells remains a daunting task, as they are actively cleared by the recipient's immune system. Therefore, a detailed understanding of the innate or adaptive immune cells-derived responses against allogeneic transplanted hepatic cells is the key to rationalize cell-based therapies. METHODS: Here, we induced an acute inflammatory regenerative niche (3-96 h) on the surface of the liver by the application of cryo-injury (CI) to systematically evaluate the innate immune response against transplanted allogeneic hepatic progenitors in a sustained micro-inflammatory environment. RESULTS: The resulting data highlighted that the injured site was significantly repopulated by alternating numbers of innate immune cells, including neutrophils, monocytes and Kupffer cells (KCs), from 3 to 96 h. The transplanted allo-HPs, engrafted 6 h post-injury, were collectively eliminated by the innate immune response within 24 h of transplantation. Selective depletion of the KCs demonstrated a delayed recruitment of monocytes from day 2 to day 6. In addition, the intrasplenic engraftment of the hepatic progenitors 54 h post-transplantation was dismantled by KCs, while a time-dependent better survival and translocation of the transplanted cells into the injured site could be observed in samples devoid of KCs. CONCLUSION: Overall, this study provides evidence that KCs ablation enables a better survival and integration of allo-HPs in a sustained liver inflammatory environment, having implications for rationalizing the cell-based therapeutic interventions against liver defects.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Macrófagos del Hígado , Macrófagos del Hígado/fisiología , Hígado , Hepatocitos/trasplante , Regeneración Hepática/fisiología
3.
Cell Stem Cell ; 30(3): 283-299.e9, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36787740

RESUMEN

Stem cell-independent reprogramming of differentiated cells has recently been identified as an important paradigm for repairing injured tissues. Following periportal injury, mature hepatocytes re-activate reprogramming/progenitor-related genes (RRGs) and dedifferentiate into liver progenitor-like cells (LPLCs) in both mice and humans, which contribute remarkably to regeneration. However, it remains unknown which and how external factors trigger hepatocyte reprogramming. Here, by employing single-cell transcriptional profiling and lineage-specific deletion tools, we uncovered that periportal-specific LPLC formation was initiated by regionally activated Kupffer cells but not peripheral monocyte-derived macrophages. Unexpectedly, using in vivo screening, the proinflammatory factor IL-6 was identified as the niche signal repurposed for RRG induction via STAT3 activation, which drove RRG expression through binding to their pre-accessible enhancers. Notably, RRGs were activated through injury-specific rather than liver embryogenesis-related enhancers. Collectively, these findings depict an injury-specific niche signal and the inflammation-mediated transcription in driving the conversion of hepatocytes into a progenitor phenotype.


Asunto(s)
Interleucina-6 , Macrófagos del Hígado , Animales , Humanos , Ratones , Diferenciación Celular , Hepatocitos/metabolismo , Interleucina-6/metabolismo , Macrófagos del Hígado/fisiología , Hígado , Regeneración Hepática/fisiología
4.
Int Rev Cell Mol Biol ; 368: 143-212, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35636927

RESUMEN

Macrophages are a heterogeneous population of innate immune cells and key cellular components of the liver. Hepatic macrophages consist of embryologically-derived resident Kupffer cells (KC), recruited monocyte-derived macrophages (MDM) and capsular macrophages. Both the diversity and plasticity of hepatic macrophage subsets explain their different functions in the maintenance of hepatic homeostasis and in injury processes in acute and chronic liver diseases. In this review, we assess the evidence for macrophage involvement in regulating both liver health and injury responses in liver diseases including acute liver injury (ALI), chronic liver disease (CLD) (including liver fibrosis) and hepatocellular carcinoma (HCC). In healthy livers, KC display critical functions such as phagocytosis, danger signal recognition, cytokine release, antigen processing and the ability to orchestrate immune responses and maintain immunological tolerance. However, in most liver diseases there is a striking hepatic MDM expansion, which orchestrate both disease progression and regression. Single-cell approaches have transformed our understanding of liver macrophage heterogeneity, dynamics, and functions in both human samples and preclinical models. We will further discuss the new insights provided by these approaches and how they are enabling high-fidelity work to specifically identify pathogenic macrophage subpopulations. Given the important role of macrophages in regulating injury responses in a broad range of settings, there is now a huge interest in developing new therapeutic strategies aimed at targeting macrophages. Therefore, we also review the current approaches being used to modulate macrophage function in liver diseases and discuss the therapeutic potential of targeting macrophage subpopulations as a novel treatment strategy for patients with liver disorders.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/patología , Humanos , Macrófagos del Hígado/patología , Macrófagos del Hígado/fisiología , Macrófagos
5.
Toxicology ; 468: 153101, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35065160

RESUMEN

Monocrotaline (MCT), an unsaturated pyrrolizidine alkaloid (PA) in plants, is mainly toxic to the lung and liver of mammals. As a commonly used tool for liver injury model, the mechanism of MCT hepatoxicity has still not been fully clarified. Kupffer cells (KCs) are the liver-resident macrophages and have various responses to different toxicants and liver damage. However, the role of KCs in MCT-induced liver injury remains controversial. In current work, we investigated the effects of KCs on MCT-induced liver injury, especially on MCT-induced hepatocyte death. KCs were depleted in Balb/c mice by liposome-entrapped clodronate (Lip/Clo) (0.2 mL/mouse, i.p.) or GdCl3 (0.7 mg/kg, i.p.) before MCT administration (300 mg/kg, i.p.), we found that the Lip/Clo group showed higher efficiency in KCs depletion and stronger hepatoprotective effects against MCT. We also found TNF-α was remarkably decreased after KCs depletion, the experiment of administering anti-TNF-α antibody (20 µg/mouse, i.p.) to MCT-treated animals generated the similar results. To further elaborate the function of KCs, we compared the ALT levels released from co-culturing murine hepatic parenchymal cells (HPCs) and RAW264.7 cells with that from HPCs alone. After the treatment of MCT, the released ALT levels in co-culture system were shown to be dependent on the number of RAW264.7 cells, while the anti-TNF-α antibody could suppress it. Finally, we discovered RIPK3/MLKL pathway might be activated by TNF-α released from KCs, and subsequently induced hepatocyte necrosis. Noteworthy, the known mechanisms including ER stress and NF-κB pathways were also found to be involved in the activation of KCs. In conclusion, our study reveals a further mechanism to MCT-induced hepatoxicity mediated directly by KCs via producing TNF-α.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Macrófagos del Hígado/fisiología , Monocrotalina/toxicidad , Factor de Necrosis Tumoral alfa/biosíntesis , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Hemaglutininas/sangre , Masculino , Ratones , Ratones Endogámicos BALB C
6.
Surg Today ; 52(2): 344-353, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34568969

RESUMEN

AIM: To investigate the relationship between the intrahepatic expression of podoplanin (PDPN) and Kupffer cells (KCs) in ischemia-reperfusion (I/R) liver damage. METHODS: C57Bl/6 mice were injected with 200 µl of clodronate liposomes (macrophage depletion; MDP group) to deplete KCs or control liposomes (control group) via the ophthalmic vein plexus 24 h prior to ischemia. Animals were subjected to 90 min of partial hepatic ischemia (70%), followed by reperfusion, and were then killed at designated time points. Serum and liver tissues were harvested for further analyses. RESULTS: Serum ALT levels, mortality rates, and the percentage of necrotic area in liver sections were significantly higher in the MDP group than in the control group. PDPN was expressed in the lymphatic epithelium, interlobular bile duct epithelium, and in some hepatocytes in each group. Its expression in hepatocytes was down-regulated in the MDP group. The accumulation of platelets in the sinusoid was reduced 6 h after I/R in the MDP group. Tissue HGF and IGF-1 levels decreased in the MDP group. CONCLUSIONS: These results suggest that KCs play a key role in the activation of platelets through direct contact with PDPN-positive hepatocytes in I/R livers.


Asunto(s)
Isquemia/complicaciones , Macrófagos del Hígado/fisiología , Hepatopatías/etiología , Hígado/irrigación sanguínea , Hígado/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiología , Daño por Reperfusión/etiología , Alanina Transaminasa/sangre , Animales , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Hepatocitos/fisiología , Masculino , Ratones Endogámicos C57BL , Activación Plaquetaria
7.
Genes (Basel) ; 12(9)2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34573346

RESUMEN

The production of around 2.5 million red blood cells (RBCs) per second in erythropoiesis is one of the most intense activities in the body. It continuously consumes large amounts of iron, approximately 80% of which is recycled from aged erythrocytes. Therefore, similar to the "making", the "breaking" of red blood cells is also very rapid and represents one of the key processes in mammalian physiology. Under steady-state conditions, this important task is accomplished by specialized macrophages, mostly liver Kupffer cells (KCs) and splenic red pulp macrophages (RPMs). It relies to a large extent on the engulfment of red blood cells via so-called erythrophagocytosis. Surprisingly, we still understand little about the mechanistic details of the removal and processing of red blood cells by these specialized macrophages. We have only started to uncover the signaling pathways that imprint their identity, control their functions and enable their plasticity. Recent findings also identify other myeloid cell types capable of red blood cell removal and establish reciprocal cross-talk between the intensity of erythrophagocytosis and other cellular activities. Here, we aimed to review the multiple and emerging facets of iron recycling to illustrate how this exciting field of study is currently expanding.


Asunto(s)
Eritrocitos/fisiología , Hemólisis/fisiología , Hierro/metabolismo , Macrófagos/fisiología , Fagocitosis/fisiología , Animales , Eritrocitos/patología , Humanos , Macrófagos del Hígado/fisiología , Hígado/citología , Hígado/fisiología , Macrófagos/inmunología
8.
Hepatology ; 74(4): 2118-2132, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33999437

RESUMEN

BACKGROUND AND AIMS: Liver ischemia reperfusion injury (IRI) remains an unresolved clinical problem. This study dissected roles of liver-resident macrophage Kupffer cells (KCs), with a functional focus on efferocytosis receptor T-cell immunoglobulin and mucin domain-containing protein-4 (TIM-4), in both the activation and resolution of IRI in a murine liver partial warm ischemia model. APPROACH AND RESULTS: Fluorescence-activated cell sorting results showed that TIM-4 was expressed exclusively by KCs, but not infiltrating macrophages (iMФs), in IR livers. Anti-TIM-4 antibody depleted TIM-4+ macrophages in vivo, resulting in either alleviation or deterioration of liver IRI, which was determined by the repopulation kinetics of the KC niche with CD11b+ macrophages. To determine the KC-specific function of TIM-4, we reconstituted clodronate-liposome-treated mice with exogenous wild-type or TIM-4-deficient KCs at either 0 hour or 24 hours postreperfusion. TIM-4 deficiency in KCs resulted in not only increases in the severity of liver IRI (at 6 hours postreperfusion), but also impairment of the inflammation resolution (at 7 days postreperfusion). In vitro analysis revealed that TIM-4 promoted KC efferocytosis to regulate their Toll-like receptor response by up-regulating IL-10 and down-regulating TNF-α productions. CONCLUSIONS: TIM-4 is critical for KC homeostatic function in both the activation and resolution of liver IRI by efferocytosis.


Asunto(s)
Interleucina-10/metabolismo , Macrófagos del Hígado/fisiología , Hepatopatías/inmunología , Proteínas de la Membrana/metabolismo , Daño por Reperfusión/inmunología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Homeostasis/fisiología , Inflamación/metabolismo , Ratones , Transducción de Señal
9.
Int J Cancer ; 148(5): 1276-1288, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33038274

RESUMEN

The liver ischemia-reperfusion (IR) injury that occurs consequently to hepatic resection performed in patients with metastases can lead to tumor relapse for not fully understood reasons. We assessed the effects of liver IR on tumor growth and the innate immune response in a mouse model of colorectal (CR) liver metastasis. Mice subjected to liver ischemia 2 days after intrasplenic injection of CR carcinoma cells displayed a higher metastatic load in the liver, correlating with Kupffer cells (KC) death through the activation of receptor-interating protein 3 kinase (RIPK3) and caspase-1 and a recruitment of monocytes. Interestingly, the immunoregulatory mediators, tumor necrosis factor-α (TNF-α) and heme oxygenase-1 (HO-1) were strongly upregulated in recruited monocytes and were also expressed in the surviving KC following IR. Using TNFflox/flox LysMcre/wt mice, we showed that TNF deficiency in macrophages and monocytes favors tumor progression after IR. The antitumor effect of myeloid cell-derived TNF involved direct tumor cell apoptosis and a reduced expression of immunosuppressive molecules such as transforming growth factor-ß, interleukin (IL)-10, inducible nitric oxyde synthase (iNOS), IL-33 and HO-1. Conversely, a monocyte/macrophage-specific deficiency in HO-1 (HO-1flox/flox LysMcre/wt ) or the blockade of HO-1 function led to the control of tumor progression post-liver IR. Importantly, host cell RIPK3 deficiency maintains the KC number upon IR, inhibits the IR-induced innate cell recruitment, increases the TNF level, decreases the HO-1 level and suppresses the tumor outgrowth. In conclusion, tumor recurrence in host undergoing liver IR is associated with the death of antitumoral KC and the recruitment of monocytes endowed with immunosuppressive properties. In both of which HO-1 inhibition would reinforce their antitumoral activity.


Asunto(s)
Neoplasias Colorrectales/patología , Hemo-Oxigenasa 1/fisiología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/secundario , Hígado/irrigación sanguínea , Recurrencia Local de Neoplasia/etiología , Daño por Reperfusión/complicaciones , Factor de Necrosis Tumoral alfa/fisiología , Animales , Progresión de la Enfermedad , Macrófagos del Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología
10.
Cells ; 9(10)2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33050035

RESUMEN

A high fat Western-style diet leads to hepatic steatosis that can progress to steatohepatitis and ultimately cirrhosis or liver cancer. The mechanism that leads to the development of steatosis upon nutritional overload is complex and only partially understood. Using click chemistry-based metabolic tracing and microscopy, we study the interaction between Kupffer cells and hepatocytes ex vivo. In the early phase of steatosis, hepatocytes alone do not display significant deviations in fatty acid metabolism. However, in co-cultures or supernatant transfer experiments, we show that tumor necrosis factor (TNF) secretion by Kupffer cells is necessary and sufficient to induce steatosis in hepatocytes, independent of the challenge of hepatocytes with elevated fatty acid levels. We further show that free fatty acid (FFA) or lipopolysaccharide are both able to trigger release of TNF from Kupffer cells. We conclude that Kupffer cells act as the primary sensor for both FFA overload and bacterial lipopolysaccharide, integrate these signals and transmit the information to the hepatocyte via TNF secretion. Hepatocytes react by alteration in lipid metabolism prominently leading to the accumulation of triacylglycerols (TAGs) in lipid droplets, a hallmark of steatosis.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Hepatocitos/metabolismo , Macrófagos del Hígado/metabolismo , Animales , Química Clic/métodos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/fisiología , Hígado Graso/etiología , Hígado Graso/metabolismo , Hepatocitos/fisiología , Inflamación/metabolismo , Macrófagos del Hígado/fisiología , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factor de Necrosis Tumoral alfa
11.
Biomed Pharmacother ; 131: 110693, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32882586

RESUMEN

Macrophages play a crucial role in the pathogenesis of pancreatitis that is a common gastrointestinal disease. Particularly, macrophages differentiate into different phenotypes and exert diverse functions in acute pancreatitis (AP) and chronic pancreatitis (CP), respectively. In AP, macrophages in the pancreas and other related organs are mainly activated and differentiated into a pro-inflammatory M1 phenotype, and furthermore secrete inflammatory cytokines and mediators, causing local inflammation of the pancreas, and even intractable systemic inflammatory response or multiple organ failure. In CP, macrophages often exhibit a M2 polarisation and interact with pancreatic stellate cells (PSCs) in an autocrine and paracrine cytokine-dependent manner to promote the progression of pancreatic fibrosis. As the severity of pancreatic fibrosis aggravates, the proportion of M2/M1 macrophage cytokines in the pancreas increases. The discovery of macrophages in the pathogenesis of pancreatitis has promoted the research of targeted drugs, which provides great potential for the effective treatment of pancreatitis. This paper provides an overview of the roles of various macrophages in the pathogenesis of pancreatitis and the current research status of pancreatitis immunotherapy targeting macrophages. The findings addressed in this review are of considerable significance for understanding the pivotal role of macrophages in pancreatitis.


Asunto(s)
Macrófagos/fisiología , Pancreatitis/etiología , Animales , Humanos , Inmunoterapia , Macrófagos del Hígado/fisiología , Pancreatitis/tratamiento farmacológico , Pancreatitis Crónica/tratamiento farmacológico , Pancreatitis Crónica/etiología
12.
Methods Mol Biol ; 2164: 65-73, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32607884

RESUMEN

Here, we describe a protocol to prepare and administer glucan-encapsulated RNAi particles (GeRPs), for specific delivery of siRNA and subsequent gene silencing in Kupffer cells (KCs) in mice. This technology is based on baker's yeast and allows gene manipulation in macrophages in a tissue-specific manner depending on the route of administration and the model that is used. GeRP administered by intravenous injection in mice are delivered to KCs. Therefore, using the GeRP technology to silence genes provides a unique method to study the function of factors expressed by KCs in the regulation of liver function.


Asunto(s)
Silenciador del Gen/fisiología , Glucanos/genética , Macrófagos del Hígado/fisiología , Hígado/fisiología , ARN Interferente Pequeño/genética , Animales , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN/fisiología
13.
Front Immunol ; 11: 322, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32362892

RESUMEN

Ample evidence suggests that hepatic macrophages play key roles in the injury and repair mechanisms during liver disease progression. There are two major populations of hepatic macrophages: the liver resident Kupffer cells and the monocyte-derived macrophages, which rapidly infiltrate the liver during injury. Under different disease conditions, the tissue microenvironmental cues of the liver critically influence the phenotypes and functions of hepatic macrophages. Furthermore, hepatic macrophages interact with multiple cells types in the liver, such as hepatocytes, neutrophils, endothelial cells, and platelets. These crosstalk interactions are of paramount importance in regulating the extents of liver injury, repair, and ultimately liver disease progression. In this review, we summarize the novel findings highlighting the impact of injury-induced microenvironmental signals that determine the phenotype and function of hepatic macrophages. Moreover, we discuss the role of hepatic macrophages in homeostasis and pathological conditions through crosstalk interactions with other cells of the liver.


Asunto(s)
Comunicación Celular , Macrófagos del Hígado/fisiología , Hígado/patología , Macrófagos/fisiología , Hepatocitos/fisiología , Homeostasis , Humanos , Células T Asesinas Naturales/fisiología , Fagocitosis
14.
Nat Commun ; 11(1): 1269, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32152267

RESUMEN

Multicellular rosettes are transient epithelial structures that serve as intermediates during diverse organ formation. We have identified a unique contributor to rosette formation in zebrafish Kupffer's vesicle (KV) that requires cell division, specifically the final stage of mitosis termed abscission. KV utilizes a rosette as a prerequisite before forming a lumen surrounded by ciliated epithelial cells. Our studies identify that KV-destined cells remain interconnected by cytokinetic bridges that position at the rosette's center. These bridges act as a landmark for directed Rab11 vesicle motility to deliver an essential cargo for lumen formation, CFTR (cystic fibrosis transmembrane conductance regulator). Here we report that premature bridge cleavage through laser ablation or inhibiting abscission using optogenetic clustering of Rab11 result in disrupted lumen formation. We present a model in which KV mitotic cells strategically place their cytokinetic bridges at the rosette center, where Rab11-associated vesicles transport CFTR to aid in lumen establishment.


Asunto(s)
División Celular/fisiología , Polaridad Celular/fisiología , Embrión no Mamífero/fisiología , Desarrollo Embrionario/fisiología , Macrófagos del Hígado/fisiología , Organogénesis/fisiología , Pez Cebra/embriología , Animales , Línea Celular , Movimiento Celular , Cilios/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Macrófagos del Hígado/citología , Mitosis , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
15.
Front Immunol ; 11: 609618, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33574817

RESUMEN

Kupffer cells, the resident macrophages of the liver, comprise the largest pool of tissue macrophages in the body. Within the liver sinusoids Kupffer cells perform functions common across many tissue macrophages including response to tissue damage and antigen presentation. They also engage in specialized activities including iron scavenging and the uptake of opsonized particles from the portal blood. Here, we review recent studies of the epigenetic pathways that establish Kupffer cell identity and function. We describe a model by which liver-environment specific signals induce lineage determining transcription factors necessary for differentiation of Kupffer cells from bone-marrow derived monocytes. We conclude by discussing how these lineage determining transcription factors (LDTFs) drive Kupffer cell behavior during both homeostasis and disease, with particular focus on the relevance of Kupffer cell LDTF pathways in the setting of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis.


Asunto(s)
Epigénesis Genética/genética , Hígado Graso/genética , Macrófagos del Hígado/patología , Macrófagos del Hígado/fisiología , Enfermedad del Hígado Graso no Alcohólico/genética , Animales , Médula Ósea/patología , Médula Ósea/fisiología , Diferenciación Celular/genética , Hígado Graso/patología , Homeostasis/genética , Humanos , Hígado/patología , Hígado/fisiología , Macrófagos/patología , Macrófagos/fisiología , Monocitos/patología , Monocitos/fisiología , Enfermedad del Hígado Graso no Alcohólico/patología , Transducción de Señal/genética
16.
J Gastroenterol Hepatol ; 35(6): 960-966, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31782974

RESUMEN

Performance and advances in liver surgery makes remarkable progress of the understanding of liver regeneration. Liver regeneration after liver resection has been widely researched, and the underlying mechanism mostly concerns proliferation of hepatocytes and the influence by inflammation through activation of Kupffer cells and the other parenchymal cells, the second regenerative pathway by hepatic progenitor cells (HPCs), inducing angiogenesis, remodeling of a extracellular matrix (ECM), and termination mechanisms. New clinical surgeries and the updated multiomics analysis are exploiting the remarkable progress, especially in immune regulation and metabolic process of two emerging hallmarks. This review briefly represents a systemic outline of eight hallmarks, including hepatocyte proliferation, contribution of hepatic progenitor cells, inducing angiogenesis, reprogramming of the extracellular matrix, apoptosis and termination of proliferation, inflammation, immune and metabolic regulation, which are set as organizing characteristics of postoperative liver regeneration and future directions of refining treatment targets.


Asunto(s)
Procedimientos Quirúrgicos del Sistema Digestivo , Regeneración Hepática/fisiología , Hígado/cirugía , Proliferación Celular , Matriz Extracelular/metabolismo , Hepatocitos/fisiología , Humanos , Macrófagos del Hígado/fisiología , Hígado/citología , Hígado/metabolismo , Hígado/fisiopatología , Neovascularización Fisiológica , Tejido Parenquimatoso/citología , Tejido Parenquimatoso/fisiología , Periodo Posoperatorio , Células Madre/fisiología
17.
Immunity ; 51(4): 655-670.e8, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31587991

RESUMEN

Tissue environment plays a powerful role in establishing and maintaining the distinct phenotypes of resident macrophages, but the underlying molecular mechanisms remain poorly understood. Here, we characterized transcriptomic and epigenetic changes in repopulating liver macrophages following acute Kupffer cell depletion as a means to infer signaling pathways and transcription factors that promote Kupffer cell differentiation. We obtained evidence that combinatorial interactions of the Notch ligand DLL4 and transforming growth factor-b (TGF-ß) family ligands produced by sinusoidal endothelial cells and endogenous LXR ligands were required for the induction and maintenance of Kupffer cell identity. DLL4 regulation of the Notch transcriptional effector RBPJ activated poised enhancers to rapidly induce LXRα and other Kupffer cell lineage-determining factors. These factors in turn reprogrammed the repopulating liver macrophage enhancer landscape to converge on that of the original resident Kupffer cells. Collectively, these findings provide a framework for understanding how macrophage progenitor cells acquire tissue-specific phenotypes.


Asunto(s)
Macrófagos del Hígado/fisiología , Hígado/metabolismo , Macrófagos/fisiología , Células Mieloides/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Microambiente Celular , Reprogramación Celular , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/citología , Receptores X del Hígado/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
18.
Immunity ; 51(4): 638-654.e9, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31561945

RESUMEN

Macrophages are strongly adapted to their tissue of residence. Yet, little is known about the cell-cell interactions that imprint the tissue-specific identities of macrophages in their respective niches. Using conditional depletion of liver Kupffer cells, we traced the developmental stages of monocytes differentiating into Kupffer cells and mapped the cellular interactions imprinting the Kupffer cell identity. Kupffer cell loss induced tumor necrosis factor (TNF)- and interleukin-1 (IL-1) receptor-dependent activation of stellate cells and endothelial cells, resulting in the transient production of chemokines and adhesion molecules orchestrating monocyte engraftment. Engrafted circulating monocytes transmigrated into the perisinusoidal space and acquired the liver-associated transcription factors inhibitor of DNA 3 (ID3) and liver X receptor-α (LXR-α). Coordinated interactions with hepatocytes induced ID3 expression, whereas endothelial cells and stellate cells induced LXR-α via a synergistic NOTCH-BMP pathway. This study shows that the Kupffer cell niche is composed of stellate cells, hepatocytes, and endothelial cells that together imprint the liver-specific macrophage identity.


Asunto(s)
Células Endoteliales/fisiología , Células Estrelladas Hepáticas/fisiología , Hepatocitos/fisiología , Macrófagos del Hígado/fisiología , Hígado/citología , Macrófagos/fisiología , Monocitos/fisiología , Animales , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Microambiente Celular , Femenino , Regulación de la Expresión Génica , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/metabolismo
19.
IUBMB Life ; 71(12): 1952-1961, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31408278

RESUMEN

This study aims to investigate the role of long noncoding RNA SNHG20 in the progression of nonalcoholic fatty liver disease (NALFD) to hepatocellular carcinoma (HCC), and to elucidate whether polarization of Kupffer cells (KCs, liver macrophages) was involved in this process. Mouse NALFD was induced by 16 weeks of high-fat diet (HFD) feeding. Mouse NALFD-HCC was induced by 36 weeks of HFD feeding (from 1 week to 36 weeks) and 20 weeks of diethyl nitrosamine (DEN) administration (from 17 weeks to 36 weeks). The LV-shRNA- and LV-sh-SNHG20-infected RAW264.7 cells were injected into the NALFD mice followed by DEN treatment to evaluate the role of SNHG20 in regulating the progression of NALFD to HCC in mice. The proportion of M1 and M2 macrophages was determined by flow cytometry. The levels of M1-related inducible nitric oxide synthase (iNOS) and tumor necrosis factor-α and M2-related Arg-1 and interleukin (IL)-10 were also examined. SNHG20 expression was decreased in NALFD but increased in NALFD-HCC, both in human and experimental mouse livers. Furthermore, human and mouse NALFD-HCC KCs displayed decreased M1/M2 ratio compared with NALFD KCs. Moreover, SNHG20 overexpression induced M2 polarization through activating STAT6, whereas SNHG20 silencing suppressed M1 polarization in RAW264.7 macrophages and delayed the progression of NALFD to HCC in mice. SNHG20 may facilitate the progression of NALFD to HCC via inducing liver KCs M2 polarization via STAT6 activation.


Asunto(s)
Macrófagos del Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Largo no Codificante/genética , Animales , Carcinoma Hepatocelular/patología , Polaridad Celular/genética , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Macrófagos del Hígado/fisiología , Hígado/patología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo
20.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31160364

RESUMEN

Pneumonia and sepsis are distinct but integrally linked public health concerns. The hepatic acute-phase response (APR), which is largely dependent on transcription factors NF-κB RelA and STAT3, is a hallmark of these pathologies and other injurious conditions. Inactivation of the APR can promote liver injury, a frequently observed organ dysfunction during sepsis. However, whether or how the acute-phase changes promote liver tissue resilience during infections is unclear. To determine the hepatoprotective role of the hepatic APR, we utilized mice bearing hepatocyte-specific deletions of either RelA or STAT3. Mice were challenged intratracheally (i.t.), intravenously (i.v.), or intraperitoneally (i.p.) with Escherichia coli, Klebsiella pneumoniae, Streptococcus pneumoniae, lipopolysaccharide (LPS), or alpha-galactosylceramide (αGalCer) to induce pneumonia, sepsis, or NKT cell activation. Liver injury was observed in RelA-null (hepRelAΔ/Δ) mice but not STAT3-null (hepSTAT3Δ/Δ) mice during pneumonia. The absence of RelA resulted in hepatotoxicity across several models of pneumonia, sepsis, and NKT cell activation. Injury was associated with increased levels of activated caspase-3 and -8 and substantial alteration of the hepatic transcriptome. Hepatotoxicity in the absence of RelA could be reversed by neutralization of tumor necrosis factor alpha (TNF-α). These results indicate the requirement of RelA-dependent inducible hepatoprotection during pneumonia and sepsis. Further, the results demonstrate that RelA-dependent gene programs are critical for maintaining liver homeostasis against TNF-α-driven immunotoxicity.


Asunto(s)
Hígado/patología , Neumonía/patología , Sepsis/patología , Factor de Transcripción ReIA/fisiología , Reacción de Fase Aguda , Animales , Apoptosis , Quimiocina CCL2/fisiología , Macrófagos del Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , Células T Asesinas Naturales/inmunología , Factor de Transcripción STAT3/fisiología , Factor de Necrosis Tumoral alfa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA