Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Visc Med ; 39(6): 163-165, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38205273
2.
J Crohns Colitis ; 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38285546

RESUMEN

BACKGROUND: HMGB1 is a ubiquitous nucleoprotein with immune-regulatory properties following cellular secretion or release in sterile and infectious inflammation. Stool and serum HMGB1 levels correlate with colitis severity and colorectal cancer (CRC) progression, yet recent reports indicated HMGB1 to mainly operate as an intracellular determinant of enterocyte fate during colitis, and investigations into the roles of HMGB1 in CRC are lacking. Using mice with conditional HMGB1-knockout in enterocytes (Hmgb1ΔIEC) and myeloid cells (Hmgb1ΔLysM), respectively, we explored functions of HMGB1 in pathogenetically diverse contexts of colitis and colitis-associated CRC. RESULTS: HMGB1 is overexpressed in human inflammatory bowel disease and gastrointestinal cancers, and HMGB1 protein localizes in enterocytes and stromal cells in colitis and CRC specimens from humans and rodents. As previously described, enterocyte HMGB1 deficiency aggravates severe chemical-induced intestinal injury, but not Citrobacter rodentium or T cell transfer colitis in mice. HMGB1-deficient enterocytes and organoids do not exhibit deviant apoptotic or autophagic activity, altered proliferative or migratory capacity, abnormal intestinal permeability or aberrant DSS-induced organoid inflammation in vitro. Instead, we observed altered in vivo-reprogramming of both intestinal epithelia and infiltrating myeloid cells in Hmgb1ΔIEC early during colitis, suggesting HMGB1-mediated paracrine injury signaling. Hmgb1ΔIEC had higher CRC burden than wildtypes in the Apc+/min model, whereas inflammatory CRC was attenuated in Hmgb1ΔLysM. Cellular and molecular phenotyping of Hmgb1ΔIEC and Hmgb1ΔLysM cancers indicates context-dependent transcriptional modulation of immune signaling and extracellular matrix remodeling via HMGB1. CONCLUSION: Enterocytes and myeloid cells context-dependently regulate host responses to severe colitis and maladaptive intestinal wound healing via HMGB1.

3.
World J Hepatol ; 15(12): 1325-1332, 2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38223419

RESUMEN

BACKGROUND: Periodontitis has been associated with various liver diseases. However, the relevance of periodontitis in the progression of decompensated cirrhosis remains inconclusive. In particular, it is unclear whether the common periodontitis pathogens, Porphyromonas gingivalis (P. gingivalis) and Actinobacillus actinomycetemcomitans (A. actinomycetemcomitans), can be detected not only in the oral mucosa but also in ascites and stool. AIM: To investigate the significance of periodontitis, P. gingivalis, and A. actinomycetemcomitans in cirrhosis patients with ascitic decompensation. METHODS: This prospective study was conducted at the University Hospital Hamburg-Eppendorf, a tertiary center in Northern Germany. A cohort of 27 patients with ascitic decompensated liver cirrhosis underwent dental examinations to assess the association between periodontitis and various clinical parameters of cirrhosis, as well as patient outcomes. PCR was used to test gingival samples, ascites, and stool for the presence of P. gingivalis and A. actinomycetemcomitans. Gingival samples were collected by probing the deepest gum pocket of a sextant and wiping them on a cotton swab. RESULTS: Periodontitis was diagnosed in 22 out of 27 (82%) ascite patients, which is significantly more common than in a control cohort of 100 unselected patients (59%, P = 0.04). P. gingivalis was detected in the gingiva of six patients, and one of them also had P. gingivalis in their stool. However, P. gingivalis was not found in the ascites of any patient. Five out of six patients with P. gingivalis had periodontitis (83%). A. actinomycetemcomitans was not detected in any sample. Patients without periodontitis had a significantly higher mortality rate compared to those with periodontitis, and survival (Kaplan-Meier analysis) was longer in patients with periodontitis (P = 0.02). Transplant-free survival was also more common in patients with periodontitis compared to those without (63% vs 0%, P = 0.02). CONCLUSION: Decompensated cirrhotic patients frequently suffer from periodontitis. However, there was no evidence of the translocation of P. gingivalis or A. actinomycetemcomitans into ascites. The survival of cirrhotic patients with periodontitis was not reduced.

4.
Eur J Intern Med ; 102: 54-62, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35672219

RESUMEN

BACKGROUND & AIMS: Acute pancreatitis (AP) is a frequent indication for hospitalization and may present with varying degrees of severity. AP often coincides with hepatic disease, yet the impact of liver cirrhosis (LC) on the course of AP is uncertain, and early identification of patients at risk for complications remains challenging. We aimed to assess the impact of LC on the development of pancreatic and extra-pancreatic complications of AP, and to identify predictors of adverse outcomes in cirrhotic patients. METHODS: All adult patients with LC and AP (LC-AP, n = 52) admitted to our institution between 01/2011-03/2020 were subjected to a 1:2 matched-pair analysis with patients with AP but without LC (NLC-AP, n = 104). RESULTS: At hospital admission, Glasgow-Imrie and Ranson scores as well as markers of systemic inflammation were comparable in LC-AP and NLC-AP patients, and both groups had similar rates of necrotizing AP. Infectious complications were more prevalent, and medical interventions were performed more often and with higher complication rates in LC-AP patients. While only 12.5% of NLC-AP patients developed organ failures, 48% of LC-AP patients developed single (7.7%) or multiple organ failure (40.4%), resulting in 44% of LC-AP patients with acute-on-chronic liver failure (ACLF). Patients with overt portal hypertension were particularly prone for decompensation. Mortality was higher among LC-AP compared to NLC-AP patients (6-month mortality 25% vs. 1.9%, p < 0.001), and SOFA and MELD scores at admission most accurately predicted outcomes in LC-AP. CONCLUSION: Among AP patients, concomitant cirrhosis substantially increases the risk for infections, periprocedural complications, multiorgan failure and death.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada , Pancreatitis , Enfermedad Aguda , Insuficiencia Hepática Crónica Agudizada/complicaciones , Adulto , Humanos , Cirrosis Hepática/complicaciones , Insuficiencia Multiorgánica/complicaciones , Pancreatitis/complicaciones , Pronóstico , Estudios Prospectivos , Índice de Severidad de la Enfermedad
5.
Sci Transl Med ; 14(639): eabe5795, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35385339

RESUMEN

Fibrosis contributes to ~45% of deaths in western countries. In chronic liver disease, fibrosis is a major factor determining outcomes, but efficient antifibrotic therapies are lacking. Although platelet-derived growth factor and transforming growth factor-ß constitute key fibrogenic mediators, they do not account for the well-established link between cell death and fibrosis in the liver. Here, we hypothesized that damage-associated molecular patterns (DAMPs) may link epithelial cell death to fibrogenesis in the injured liver. DAMP receptor screening identified purinergic receptor P2Y14 among several candidates as highly enriched in hepatic stellate cells (HSCs), the main fibrogenic cell type of the liver. Conversely, P2Y14 ligands uridine 5'-diphosphate (UDP)-glucose and UDP-galactose were enriched in hepatocytes and were released upon different modes of cell death. Accordingly, ligand-receptor interaction analysis that combined proteomic and single-cell RNA sequencing data revealed P2Y14 ligands and P2Y14 receptor as a link between dying cells and HSCs, respectively. Treatment with P2Y14 ligands or coculture with dying hepatocytes promoted HSC activation in a P2Y14-dependent manner. P2Y14 ligands activated extracellular signal-regulated kinase (ERK) and Yes-associated protein (YAP) signaling in HSCs, resulting in ERK-dependent HSC activation. Global and HSC-selective P2Y14 deficiency attenuated liver fibrosis in multiple mouse models of liver injury. Functional expression of P2Y14 was confirmed in healthy and diseased human liver and human HSCs. In conclusion, P2Y14 ligands and their receptor constitute a profibrogenic DAMP pathway that directly links cell death to fibrogenesis.


Asunto(s)
Células Estrelladas Hepáticas , Hepatocitos , Receptores Purinérgicos P2Y , Receptores Purinérgicos P2 , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Humanos , Ligandos , Hígado/metabolismo , Cirrosis Hepática/patología , Ratones , Proteómica , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Análisis de la Célula Individual , Uridina Difosfato/metabolismo , Proteínas Señalizadoras YAP
6.
Liver Int ; 41(3): 574-584, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-34542228

RESUMEN

BACKGROUND: Among patients with cirrhosis, candidate selection and timing of liver transplantation (LT) remain problematic. Acute-on-chronic liver failure (ACLF) is a severe complication of cirrhosis with excessive short-term mortality rates under conservative therapeutic measures. The role of LT in the management of ACLF is uncertain. OBJECTIVE: To assess the impact of ACLF on post-LT survival and long-term graft function, morbidity and quality of life (QoL). METHODS: We retrospectively analysed all cirrhosis patients undergoing LT at our institution between 01/2009 and 12/2014. Median follow-up was 8.7 years. Long-term LT survivors were interviewed with established QoL questionnaires. RESULTS: Of 250 LT recipients, 98 fulfilled the EASL diagnostic ACLF criteria before LT ('ACLF-LT'). ACLF associated with reduced post-LT survival (HR for 6-month survival compared to non-ACLF-LT: 0.18; HR for 10-year-survival: 0.47; both P < .001) depending on ACLF severity before LT, and mainly inferred by infections both in the early and late phases after LT. In ACLF patients, CLIFc-OFs was superior to MELD score in predicting post-LT mortality. Long-term follow-up revealed comparable graft functions and comorbidity burden in ACLF-LT and non-ACLF-LT survivors. ACLF-LT patients reported significantly impaired health and QoL, particularly with regards to anxiety/depression and physical and psychological health (all P < .05). LabMELD score, presence of ACLF at LT and duration of post-LT intensive care associated with poor long-term QoL. CONCLUSION: ACLF predicts impaired post-LT survival. While long-term graft function and extrahepatic comorbidities are comparable in ACLF and non-ACLF LT survivors, the strikingly low QoL in many ACLF-LT recipients warrants consideration during follow-up patient care.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada , Trasplante de Hígado , Humanos , Cirrosis Hepática , Pronóstico , Calidad de Vida , Estudios Retrospectivos
7.
Hepatol Commun ; 5(12): 2104-2120, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34558858

RESUMEN

High-mobility group box 1 (HMGB1) is a nucleoprotein with proinflammatory functions following cellular release during tissue damage. Moreover, antibody-mediated HMGB1 neutralization alleviates lipopolysaccharide (LPS)-induced shock, suggesting a role for HMGB1 as a superordinate therapeutic target for inflammatory and infectious diseases. Recent genetic studies have indicated cell-intrinsic functions of HMGB1 in phagocytes as critical elements of immune responses to infections, yet the role of extracellular HMGB1 signaling in this context remains elusive. We performed antibody-mediated and genetic HMGB1 deletion studies accompanied by in vitro experiments to discern context-dependent cellular sources and functions of extracellular HMGB1 during murine bloodstream infection with Listeria monocytogenes. Antibody-mediated neutralization of extracellular HMGB1 favors bacterial dissemination and hepatic inflammation in mice. Hepatocyte HMGB1, a key driver of postnecrotic inflammation in the liver, does not affect Listeria-induced inflammation or mortality. While we confirm that leukocyte HMGB1 deficiency effectuates disseminated listeriosis, we observed no evidence of dysfunctional autophagy, xenophagy, intracellular bacterial degradation, or inflammatory gene induction in primary HMGB1-deficient phagocytes or altered immune responses to LPS administration. Instead, we demonstrate that mice devoid of leukocyte HMGB1 exhibit impaired hepatic recruitment of inflammatory monocytes early during listeriosis, resulting in alterations of the transcriptional hepatic immune response and insufficient control of bacterial dissemination. Bone marrow chimera indicate that HMGB1 from both liver-resident and circulating immune cells contributes to effective pathogen control. Conclusion: Leukocyte-derived extracellular HMGB1 is a critical cofactor in the immunologic control of bloodstream listeriosis. HMGB1 neutralization strategies preclude an efficient host immune response against Listeria.


Asunto(s)
Proteína HMGB1/inmunología , Inmunidad/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Sepsis/inmunología , Animales , Modelos Animales de Enfermedad , Leucocitos/inmunología , Listeriosis/microbiología , Hígado/inmunología , Hígado/microbiología , Ratones , Sepsis/microbiología , Transducción de Señal/inmunología
8.
Cell Rep ; 29(9): 2689-2701.e4, 2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31775038

RESUMEN

Regenerative responses predispose tissues to tumor formation by largely unknown mechanisms. High-mobility group box 1 (HMGB1) is a danger-associated molecular pattern contributing to inflammatory pathologies. We show that HMGB1 derived from keratinocytes, but not myeloid cells, delays cutaneous wound healing and drives tumor formation. In wounds of mice lacking HMGB1 selectively in keratinocytes, a marked reduction in neutrophil extracellular trap (NET) formation is observed. Pharmacological targeting of HMGB1 or NETs prevents skin tumorigenesis and accelerates wound regeneration. HMGB1-dependent NET formation and skin tumorigenesis is orchestrated by tumor necrosis factor (TNF) and requires RIPK1 kinase activity. NETs are present in the microenvironment of keratinocyte-derived tumors in mice and lesional and tumor skin of patients suffering from recessive dystrophic epidermolysis bullosa, a disease in which skin blistering predisposes to tumorigenesis. We conclude that tumorigenicity of the wound microenvironment depends on epithelial-derived HMGB1 regulating NET formation, thereby establishing a mechanism linking reparative inflammation to tumor initiation.


Asunto(s)
Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Piel/patología , Proteína HMGB1/metabolismo , Humanos , Microambiente Tumoral , Cicatrización de Heridas
13.
J Clin Invest ; 128(6): 2436-2451, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29558367

RESUMEN

Cell death is a key driver of disease progression and carcinogenesis in chronic liver disease (CLD), highlighted by the well-established clinical correlation between hepatocellular death and risk for the development of cirrhosis and hepatocellular carcinoma (HCC). Moreover, hepatocellular death is sufficient to trigger fibrosis and HCC in mice. However, the pathways through which cell death drives CLD progression remain elusive. Here, we tested the hypothesis that high-mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) with key roles in acute liver injury, may link cell death to injury responses and hepatocarcinogenesis in CLD. While liver-specific HMGB1 deficiency did not significantly affect chronic injury responses such as fibrosis, regeneration, and inflammation, it inhibited ductular/progenitor cell expansion and hepatocyte metaplasia. HMGB1 promoted ductular expansion independently of active secretion in a nonautonomous fashion, consistent with its role as a DAMP. Liver-specific HMGB1 deficiency reduced HCC development in 3 mouse models of chronic injury but not in a model lacking chronic liver injury. As with CLD, HMGB1 ablation reduced the expression of progenitor and oncofetal markers, a key determinant of HCC aggressiveness, in tumors. In summary, HMGB1 links hepatocyte death to ductular reaction, progenitor signature, and hepatocarcinogenesis in CLD.


Asunto(s)
Carcinoma Hepatocelular , Proteína HMGB1 , Hepatocitos , Cirrosis Hepática , Neoplasias Hepáticas , Proteínas de Neoplasias , Células Madre Neoplásicas , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Preescolar , Enfermedad Crónica , Femenino , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/lesiones , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
14.
PLoS One ; 10(10): e0140525, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26489083

RESUMEN

BACKGROUND & AIMS: Current guidelines recommend immunosuppressive treatment (IT) in patients with primary sclerosing cholangitis (PSC) and elevated aminotransferase levels more than five times the upper limit of normal and elevated serum IgG-levels above twice the upper limit of normal. Since there is no evidence to support this recommendation, we aimed to assess the criteria that guided clinicians in clinical practice to initiate IT in patients with previously diagnosed PSC. METHODS: This is a retrospective analysis of 196 PSC patients from seven German hepatology centers, of whom 36 patients had received IT solely for their liver disease during the course of PSC. Analyses were carried out using methods for competing risks. RESULTS: A simplified autoimmune hepatitis (AIH) score >5 (HR of 36, p<0.0001) and a modified histological activity index (mHAI) greater than 3/18 points (HR 3.6, p = 0.0274) were associated with the initiation of IT during the course of PSC. Of note, PSC patients who subsequently received IT differed already at the time of PSC diagnosis from those patients, who did not receive IT during follow-up: they presented with increased levels of IgG (p = 0.004) and more frequently had clinical signs of cirrhosis (p = 0.0002). CONCLUSIONS: This is the first study which investigates the parameters associated with IT in patients with PSC in clinical practice. A simplified AIH score >5 and a mHAI score >3, suggesting concomitant features of AIH, influenced the decision to introduce IT during the course of PSC. In German clinical practice, the cutoffs used to guide IT may be lower than recommended by current guidelines.


Asunto(s)
Colangitis Esclerosante/terapia , Inmunoglobulina G/sangre , Terapia de Inmunosupresión/métodos , Hepatopatías/terapia , Transaminasas/metabolismo , Adulto , Colangitis Esclerosante/diagnóstico , Colangitis Esclerosante/inmunología , Estudios de Cohortes , Femenino , Hepatitis Autoinmune/diagnóstico , Hepatitis Autoinmune/patología , Humanos , Inmunosupresores/uso terapéutico , Enfermedades Inflamatorias del Intestino/complicaciones , Hígado/patología , Hepatopatías/inmunología , Masculino , Estudios Retrospectivos
16.
Autophagy ; 11(7): 1187-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121576

RESUMEN

HMGB1 (high mobility group box 1), a ubiquitously expressed DNA-binding nucleoprotein, has not only been attributed with important functions in the regulation of gene expression but is thought to function as an important damage-associated molecular pattern in the extracellular space. Recently, conditional Hmgb1 deletion strategies have been employed to overcome the perinatal mortality of global Hmgb1 deletion and to understand HMGB1 functions under disease conditions. From these studies, it has become evident that HMGB1 is not required for normal organ function. However, the different conditional ablation strategies have yielded contradictory results in some disease models. With nearly complete recombination in all transgenic mouse models, the main reason for opposite results is likely to lie within different targeting strategies. In summary, different targeting strategies need to be taken into account when interpreting HMGB1 functions, and further efforts need to be undertaken to compare these models side by side.


Asunto(s)
Autofagia , Proteína HMGB1/metabolismo , Animales , Humanos
17.
J Clin Invest ; 125(2): 539-50, 2015 02.
Artículo en Inglés | MEDLINE | ID: mdl-25562324

RESUMEN

In contrast to microbially triggered inflammation, mechanisms promoting sterile inflammation remain poorly understood. Damage-associated molecular patterns (DAMPs) are considered key inducers of sterile inflammation following cell death, but the relative contribution of specific DAMPs, including high-mobility group box 1 (HMGB1), is ill defined. Due to the postnatal lethality of Hmgb1-knockout mice, the role of HMGB1 in sterile inflammation and disease processes in vivo remains controversial. Here, using conditional ablation strategies, we have demonstrated that epithelial, but not bone marrow-derived, HMGB1 is required for sterile inflammation following injury. Epithelial HMGB1, through its receptor RAGE, triggered recruitment of neutrophils, but not macrophages, toward necrosis. In clinically relevant models of necrosis, HMGB1/RAGE-induced neutrophil recruitment mediated subsequent amplification of injury, depending on the presence of neutrophil elastase. Notably, hepatocyte-specific HMGB1 ablation resulted in 100% survival following lethal acetaminophen intoxication. In contrast to necrosis, HMGB1 ablation did not alter inflammation or mortality in response to TNF- or FAS-mediated apoptosis. In LPS-induced shock, in which HMGB1 was considered a key mediator, HMGB1 ablation did not ameliorate inflammation or lethality, despite efficient reduction of HMGB1 serum levels. Our study establishes HMGB1 as a bona fide and targetable DAMP that selectively triggers a neutrophil-mediated injury amplification loop in the setting of necrosis.


Asunto(s)
Proteína HMGB1/metabolismo , Elastasa de Leucocito/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Receptores Inmunológicos/metabolismo , Acetaminofén/efectos adversos , Acetaminofén/farmacología , Analgésicos no Narcóticos/efectos adversos , Analgésicos no Narcóticos/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Proteína HMGB1/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Elastasa de Leucocito/genética , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Necrosis/inducido químicamente , Necrosis/genética , Necrosis/metabolismo , Necrosis/patología , Neutrófilos/patología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Choque Séptico/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
18.
J Hepatol ; 62(5): 1085-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25529619

RESUMEN

BACKGROUND & AIMS: During pregnancy, acetaminophen is one of the very few medications recommended by physicians to treat fever or pain. Recent insights from epidemiological studies suggest an association between prenatal acetaminophen medication and an increased risk for development of asthma in children later in life. The underlying pathogenesis of such association is still unknown. METHODS: We aimed to develop a mouse model to provide insights into the effect of prenatal acetaminophen on maternal, fetal and adult offspring's health. The toxic effect of acetaminophen was studied in mice on 1) maternal liver; mirrored by biomarkers of liver injury, centrilobular necrosis, and infiltration of granulocytes; 2) fetal liver; reflected by the frequency of hematopoietic stem cells, and 3) postnatal health; evaluated by the severity of allergic airway inflammation among offspring. RESULTS: We observed an increased susceptibility towards acetaminophen-induced liver damage in pregnant mice compared to virgins. Moreover, hematopoietic stem cell frequency in fetal liver declined in response to acetaminophen. Furthermore, a greater severity of airway inflammation was observed in offspring of dams upon prenatal acetaminophen treatment, identified lung infiltration by leukocytes and eosinophil infiltration into the airways. CONCLUSION: Our newly developed mouse model on prenatal use of acetaminophen reflects findings from epidemiological studies in humans. The availability of this model will allow improvement in our understanding of how acetaminophen-related hepatotoxicity is operational in pregnant individuals and how an increased risk for allergic diseases in response to prenatal acetaminophen is mediated. Such insights, once available, may change the recommendations for prenatal acetaminophen use.


Asunto(s)
Acetaminofén , Asma , Enfermedad Hepática Inducida por Sustancias y Drogas , Células Madre Fetales , Efectos Tardíos de la Exposición Prenatal , Acetaminofén/administración & dosificación , Acetaminofén/efectos adversos , Adulto , Hijos Adultos , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/efectos adversos , Animales , Asma/etiología , Asma/fisiopatología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Modelos Animales de Enfermedad , Femenino , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/patología , Humanos , Inflamación/etiología , Inflamación/fisiopatología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/prevención & control , Índice de Severidad de la Enfermedad
19.
Biomed Res Int ; 2014: 124063, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24971311

RESUMEN

Repetitive brief ischemia and reperfusion (I/R) is associated with ventricular dysfunction in pathogenesis of murine ischemic cardiomyopathy and human hibernating myocardium. We investigated the role of matricellular protein osteopontin-1 (OPN) in murine model of repetitive I/R. One 15-min LAD-occlusion followed by reperfusion was performed daily over 3, 5, and 7 consecutive days in C57/Bl6 wildtype- (WT-) and OPN(-/-)-mice (n = 8/group). After echocardiography hearts were processed for histological and mRNA-studies. Cardiac fibroblasts were isolated, cultured, and stimulated with TGF- ß 1. WT-mice showed an early, strong, and cardiomyocyte-specific osteopontin-expression leading to interstitial macrophage infiltration and consecutive fibrosis after 7 days I/R in absence of myocardial infarction. In contrast, OPN(-/-)-mice showed small, nontransmural infarctions after 3 days I/R associated with significantly worse ventricular dysfunction. OPN(-/-)-mice had different expression of myocardial contractile elements and antioxidative mediators and a lower expression of chemokines during I/R. OPN(-/-)-mice showed predominant collagen deposition in macrophage-rich small infarctions. We found lower induction of tenascin-C, MMP-9, MMP-12, and TIMP-1, whereas MMP-13-expression was higher in OPN(-/-)-mice. Cultured OPN(-/-)-myofibroblasts confirmed these findings. In conclusion, osteopontin seems to modulate expression of contractile elements, antioxidative mediators, and inflammatory response and subsequently remodel in order to protect cardiomyocytes in murine ischemic cardiomyopathy.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Osteopontina/metabolismo , Tenascina/metabolismo , Animales , Cardiomiopatías/fisiopatología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo
20.
Cell Metab ; 19(3): 539-47, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24606906

RESUMEN

In vitro studies have demonstrated a critical role for high-mobility group box 1 (HMGB1) in autophagy and the autophagic clearance of dysfunctional mitochondria, resulting in severe mitochondrial fragmentation and profound disturbances of mitochondrial respiration in HMGB1-deficient cells. Here, we investigated the effects of HMGB1 deficiency on autophagy and mitochondrial function in vivo, using conditional Hmgb1 ablation in the liver and heart. Unexpectedly, deletion of Hmgb1 in hepatocytes or cardiomyocytes, two cell types with abundant mitochondria, did not alter mitochondrial structure or function, organ function, or long-term survival. Moreover, hepatic autophagy and mitophagy occurred normally in the absence of Hmgb1, and absence of Hmgb1 did not significantly affect baseline and glucocorticoid-induced hepatic gene expression. Collectively, our findings suggest that HMGB1 is dispensable for autophagy, mitochondrial quality control, the regulation of gene expression, and organ function in the adult organism.


Asunto(s)
Autofagia , Proteína HMGB1/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Glucemia/metabolismo , Metabolismo Energético , Expresión Génica , Proteína HMGB1/genética , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Fosforilación Oxidativa , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...