Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
J Neuroinflammation ; 21(1): 123, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38725082

RESUMEN

BACKGROUND: Hepatic encephalopathy (HE) is closely associated with inflammatory responses. However, as a crucial regulator of the immune and inflammatory responses, the role of leucine-rich repeat kinase 2 (LRRK2) in the pathogenesis of HE remains unraveled. Herein, we investigated this issue in thioacetamide (TAA)-induced HE following acute liver failure (ALF). METHODS: TAA-induced HE mouse models of LRRK2 wild type (WT), LRRK2 G2019S mutation (Lrrk2G2019S) and LRRK2 knockout (Lrrk2-/-) were established. A battery of neurobehavioral experiments was conducted. The biochemical indexes and pro-inflammatory cytokines were detected. The prefrontal cortex (PFC), striatum (STR), hippocampus (HIP), and liver were examined by pathology and electron microscopy. The changes of autophagy-lysosomal pathway and activity of critical Rab GTPases were analyzed. RESULTS: The Lrrk2-/--HE model reported a significantly lower survival rate than the other two models (24% vs. 48%, respectively, p < 0.05), with no difference found between the WT-HE and Lrrk2G2019S-HE groups. Compared with the other groups, after the TAA injection, the Lrrk2-/- group displayed a significant increase in ammonium and pro-inflammatory cytokines, aggravated hepatic inflammation/necrosis, decreased autophagy, and abnormal phosphorylation of lysosomal Rab10. All three models reported microglial activation, neuronal loss, disordered vesicle transmission, and damaged myelin structure. The Lrrk2-/--HE mice presented no severer neuronal injury than the other genotypes. CONCLUSIONS: LRRK2 deficiency may exacerbate TAA-induced ALF and HE in mice, in which inflammatory response is evident in the brain and aggravated in the liver. These novel findings indicate a need of sufficient clinical awareness of the adverse effects of LRRK2 inhibitors on the liver.


Asunto(s)
Encefalopatía Hepática , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Fallo Hepático Agudo , Ratones Noqueados , Tioacetamida , Animales , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Tioacetamida/toxicidad , Ratones , Encefalopatía Hepática/patología , Encefalopatía Hepática/genética , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/genética , Masculino , Ratones Endogámicos C57BL
2.
NPJ Biofilms Microbiomes ; 10(1): 13, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38396001

RESUMEN

Both gut microbiome and microRNAs (miRNAs) play a role in the development of hepatic encephalopathy (HE). However, the functional link between the microbiome and host-derived miRNAs in faeces remains poorly understood. In the present study, patients with HE had an altered gut microbiome and faecal miRNAs compared with patients with chronic hepatitis B. Transferring faeces and faecal miRNAs from patients with HE to the recipient mice aggravated thioacetamide-induced HE. Oral gavage of hsa-miR-7704, a host-derived miRNA highly enriched in faeces from patients with HE, aggravated HE in mice in a microbiome-dependent manner. Mechanistically, hsa-miR-7704 inhibited the growth and adhesion of Bifidobacterium longum by suppressing proB. B. longum and its metabolite acetate alleviated HE by inhibiting microglial activation and ammonia production. Our findings reveal the role of miRNA-microbiome axis in HE and suggest that faecal hsa-miR-7704 are potential regulators of HE progression.


Asunto(s)
Bifidobacterium longum , Encefalopatía Hepática , MicroARNs , Animales , Humanos , Ratones , Bifidobacterium longum/genética , Bifidobacterium longum/metabolismo , Heces/microbiología , Encefalopatía Hepática/genética , Encefalopatía Hepática/microbiología , MicroARNs/genética , MicroARNs/metabolismo
3.
Eur J Med Res ; 27(1): 264, 2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36424620

RESUMEN

BACKGROUND: Hepatic encephalopathy (HE) is a reversible syndrome of brain dysfunction caused by advanced liver disease. Weighted gene co-expression network analysis (WGCNA) could establish a robust co-expression network to identify the hub genes and underlying biological functions. This study was aimed to explore the potential therapeutic targets in HE by WGCNA. RESULTS: The green and brown modules were found to be significantly associated with the development of HE. Functional enrichment analyses suggested the neuroinflammation, neuroimmune, extracellular matrix (ECM), and coagulation cascade were involved in HE. CYBB and FOXO1 were calculated as hub genes, which were upregulated in the HE patients. Tamibarotene and vitamin E were suggested as possible drug candidates to alleviate HE. CONCLUSIONS: It is the first time to analyze transcriptomic data of HE by WGCNA. Our study not only promoted the current understanding of neuroinflammation in HE, but also provided the first evidence that CYBB and FOXO1 played pivotal roles in the pathogenesis of HE, which might be potential biomarkers and therapeutic targets. Tamibarotene might be a novel drug compound against HE.


Asunto(s)
Redes Reguladoras de Genes , Encefalopatía Hepática , Humanos , Perfilación de la Expresión Génica , Encefalopatía Hepática/tratamiento farmacológico , Encefalopatía Hepática/genética , Tetrahidronaftalenos
4.
Acta Histochem ; 124(7): 151960, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36202047

RESUMEN

Hepatic encephalopathy (HE), which is caused by neurotoxin agents in the liver, is a complicated condition with a variety of neurological manifestations. Recently, endocrine alterations have been more paid attention to for neurological severity in the course of HE, e.g. adrenal gland. To identify the role of adrenal gland in the context of HE, we evaluated the functional changes of adrenal gland (i.e., plasma corticosterone concentrations and histopathological changes) in mice model of HE. To dig deep into the molecular and genetic underpinnings, a comprehensive enrichment analysis for shared genes between HE and adrenal insufficiency (AI) was also performed. Our results showed a significant reduction in the level of plasma corticosterone and severe cellular necrosis in zona fasciculate of adrenal cortex, possibly indicating adrenal insufficiency. Enrichment analysis indicated four common genes, besides predicted five novel genes and some significant MicroRNAs (miRNAs) and transcription factors for both HE and AI. Couples with, several biological processes, such as DNA damage, inflammatory responses, glycolytic processes, and insulin receptor signaling pathway were predicted in both HE and AI. To sum up, data from experimental tests and bioinformatics analyses suggest that AI play an important role in the pathogenesis and progression of HE.


Asunto(s)
Insuficiencia Suprarrenal , Encefalopatía Hepática , MicroARNs , Glándulas Suprarrenales/metabolismo , Insuficiencia Suprarrenal/etiología , Insuficiencia Suprarrenal/metabolismo , Animales , Biología Computacional , Corticosterona , Modelos Animales de Enfermedad , Encefalopatía Hepática/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Neurotoxinas/metabolismo , Receptor de Insulina/metabolismo , Factores de Transcripción/metabolismo
5.
Aliment Pharmacol Ther ; 56(10): 1475-1485, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36164267

RESUMEN

BACKGROUND: There are limited data on the diagnostic accuracy of gut microbial signatures for predicting hepatic decompensation in patients with cirrhosis. AIMS: To determine whether a stool metagenome-derived signature accurately detects hepatic decompensation and mortality risk in cirrhosis secondary to non-alcoholic fatty liver disease (NAFLD) METHODS: Shotgun metagenomic sequencing was performed on faecal samples collected at study entry from a prospective cohort of adults with NAFLD-related cirrhosis. A Random Forest machine learning algorithm was utilised to identify a metagenomic signature of decompensated cirrhosis (defined by ascites, hepatic encephalopathy or variceal haemorrhage) and subsequently validated in an external cohort. A Cox proportional hazards regression model was used to examine predictors of all-cause mortality. RESULTS: In all, 25 adults with NAFLD-related cirrhosis (training cohort) were included. Among the 16 participants with decompensated cirrhosis, 33% had ascites, 56% had hepatic encephalopathy and 22% had experienced a variceal haemorrhage (not mutually exclusive). We identified a stool metagenomic signature comprising 13 discriminatory species that reliably distinguished decompensated NAFLD-related cirrhosis (diagnostic accuracy, 0.97, 95% confidence interval [CI] 0.96-0.99). Diagnostic accuracy of the 13-species signature remained high after adjustment for lactulose (area under the curve [AUC] 0.99) and rifaximin use (AUC 0.93). The discriminative ability of 13-species metagenomic signature was robust in an independent test cohort (AUC 0.95, 95% CI 0.81-1.00). The 13-species metagenomic signature (hazard ratio [HR] 1.54, 95% CI 1.10-2.15, p = 0.01) was a stronger predictor of mortality than the Model for End-Stage Liver Disease score (HR 1.25, 95% CI 1.03-1.53, p = 0.03). CONCLUSIONS: This study provides evidence for a gut metagenome-derived signature with high diagnostic accuracy for hepatic decompensation that predicts risk of mortality in NAFLD-related cirrhosis.


Asunto(s)
Enfermedad Hepática en Estado Terminal , Várices Esofágicas y Gástricas , Encefalopatía Hepática , Enfermedad del Hígado Graso no Alcohólico , Adulto , Humanos , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/genética , Encefalopatía Hepática/etiología , Encefalopatía Hepática/genética , Várices Esofágicas y Gástricas/complicaciones , Ascitis/complicaciones , Estudios Prospectivos , Enfermedad Hepática en Estado Terminal/complicaciones , Metagenoma/genética , Rifaximina , Lactulosa , Hemorragia Gastrointestinal/etiología , Índice de Severidad de la Enfermedad , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/genética , Cirrosis Hepática/complicaciones
6.
Hum Cell ; 35(4): 1060-1070, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35583799

RESUMEN

Little is known about the role of lncRNA-mRNA regulatory relationships in hepatic encephalopathy (HE). Here, we aimed to construct the potential lncRNA and mRNA interactive network in forecasting HE development in patients with liver cirrhosis using different bioinformatic analysis method. Through analyses, we found that AL137857.1 had the most connections with other mRNAs and was deemed as a hub lncRNA. It was obviously upregulated in HE patients, which was also validated by another independent dataset. GO and KEGG analyses suggested that AL137857.1 was involved in microglial cell activation, phagocytosis, cytokine biosynthetic process, interleukin-6 production and tumor necrosis factor production. In vitro experiments suggested LPS could stimulate microglia to generate AL137857.1. In addition, we found that inhibition of AL137857.1 suppressed the expression of a series of inflammatory cytokines, including IL-1, IL-6, TNF-α, Cox2 and iNOS. Conversely, AL137857.1 over-expression induced a marked increase in these factors. Finally, AL137857.1 was demonstrated to be highly associated with the ability of microglial phagocytosis. Taken together, we have constructed a lncRNA-mRNA regulatory network associated with HE and explored the biological significance of mRNAs in the network, then discovered a novel lncRNA AL137857.1 in HE that might act as a potential regulator of the downstream inflammatory cytokines.


Asunto(s)
Encefalopatía Hepática , ARN Largo no Codificante , Citocinas/genética , Encefalopatía Hepática/genética , Encefalopatía Hepática/metabolismo , Humanos , Cirrosis Hepática/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo
7.
Oxid Med Cell Longev ; 2022: 7628522, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35464767

RESUMEN

Background: Hyperammonemia can result in various neuropathologies, including sleep disturbance, memory loss, and motor dysfunction in hepatic encephalopathy. Long noncoding RNA (lncRNA) as a group of noncoding RNA longer than 200 nucleotides is emerging as a promising therapeutic target to treat diverse diseases. Although lncRNAs have been linked to the pathogenesis of various diseases, their function in hepatic encephalopathy has not yet been elucidated. Research Design and Methods. To identify the roles of lncRNAs in hepatic encephalopathy brain, we used a bile duct ligation (BDL) mouse model and examined the alteration of neuronal cell death markers and neuronal structure-related proteins in BDL mouse cortex tissue. Furthermore, analysis of the transcriptome of BDL mouse brain cortex tissues revealed several lncRNAs critical to the apoptosis and neuronal structural changes associated with hepatic encephalopathy. Results: We confirmed the roles of the lncRNAs, ZFAS1, and GAS5 as strong candidate lncRNAs to regulate neuropathologies in hepatic encephalopathy. Our data revealed the roles of lncRNAs, ZFAS1, and GAS5, on neuronal cell death and neural structure in hyperammonemia in in vivo and in vitro conditions. Conclusion: Thus, we suggest that the modulation of these lncRNAs may be beneficial for the treatment of hepatic encephalopathy.


Asunto(s)
Encefalopatía Hepática , Hiperamonemia , ARN Largo no Codificante , Animales , Conductos Biliares , Encefalopatía Hepática/genética , Encefalopatía Hepática/patología , Hiperamonemia/complicaciones , Hiperamonemia/genética , Ratones , Neuronas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
8.
Anal Biochem ; 641: 114548, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35032459

RESUMEN

Molecular alterations underlying cerebral impairment in hyperammonemic disorders such as in hepatic encephalopathy (HE) are only poorly understood. Using transcriptomics and proteomics on brains of mice with systemic hyperammonemia resulting from knockout of hepatic glutamine synthetase (LGS-KO) we identified up to 214 genes and 34 proteins whose expressions were altered in brains of LGS-KO mice in a brain region-specific way. Differentially expressed genes were enriched for those related to oxidative stress, cell proliferation, heme metabolism and others. Due to their particularly high expression changes, coactivator associated arginine methyltransferase 1 (CARM1), TROVE2 and Lipocalin-2 (LCN2) were selected for further analyses. All selected candidates were expressed by astrocytes in rodent brain and challenging cultured astrocytes with NH4Cl changed their protein and mRNA levels similar to what was found in brains of LGS-KO mice. Further functional analyses suggested a role of CARM1 for senescence, TROVE2 for RNA quality control and LCN2 for disturbed iron homeostasis in ammonia-exposed astrocytes. LCN2 protein and Trove2 mRNA were also elevated in cerebral cortex of ammonium acetate-challenged rats and in post mortem brain tissue from patients with liver cirrhosis and HE, respectively. This study identified new molecular players potentially relevant for cerebral dysfunction in HE.


Asunto(s)
Corteza Cerebral/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Encefalopatía Hepática/metabolismo , Hiperamonemia/metabolismo , Proteoma/metabolismo , Animales , Glutamato-Amoníaco Ligasa/genética , Encefalopatía Hepática/genética , Encefalopatía Hepática/fisiopatología , Hepatocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoma/genética , Proteómica , Transcriptoma
9.
FASEB J ; 36(1): e22091, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34919756

RESUMEN

Hepatoencephalopathy due to combined oxidative phosphorylation deficiency type 1 (COXPD1) is a recessive mitochondrial translation disorder caused by mutations in GFM1, a nuclear gene encoding mitochondrial elongation factor G1 (EFG1). Patients with COXPD1 typically present hepatoencephalopathy early after birth with rapid disease progression, and usually die within the first few weeks or years of life. We have generated two different mouse models: a Gfm1 knock-in (KI) harboring the p.R671C missense mutation, found in at least 10 patients who survived more than 1 year, and a Gfm1 knock-out (KO) model. Homozygous KO mice (Gfm1-/- ) were embryonically lethal, whereas homozygous KI (Gfm1R671C/R671C ) mice were viable and showed normal growth. R671C mutation in Gfm1 caused drastic reductions in the mitochondrial EFG1 protein content in different organs. Six- to eight-week-old Gfm1R671C/R671C mice showed partial reductions of in organello mitochondrial translation and respiratory complex IV enzyme activity in the liver. Compound heterozygous Gfm1R671C/- showed a more pronounced decrease of EFG1 protein in liver and brain mitochondria, as compared with Gfm1R671C/R671C mice. At 8 weeks of age, their mitochondrial translation rates were significantly reduced in both tissues. Additionally, Gfm1R671C/- mice showed combined oxidative phosphorylation deficiency (reduced complex I and IV enzyme activities in liver and brain), and blue native polyacrylamide gel electrophoresis analysis revealed lower amounts of both affected complexes. We conclude that the compound heterozygous Gfm1R671C/- mouse presents a clear dysfunctional molecular phenotype, showing impaired mitochondrial translation and combined respiratory chain dysfunction, making it a suitable animal model for the study of COXPD1.


Asunto(s)
Encefalopatía Hepática/metabolismo , Errores Innatos del Metabolismo/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/metabolismo , Mutación Missense , Fosforilación Oxidativa , Factor G de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Encefalopatía Hepática/genética , Errores Innatos del Metabolismo/genética , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Proteínas Mitocondriales/genética , Factor G de Elongación Peptídica/genética
10.
Comput Math Methods Med ; 2021: 7777699, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34938356

RESUMEN

BACKGROUNDS: Serum long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) interaction network was discovered to exert an important role in liver cirrhosis while little is known in mild hepatic encephalopathy (MHE). Therefore, we aim to systematically evaluate the serum lncRNA-mRNA network and its regulatory mechanism in MHE. METHODS: The data of serum mRNAs and lncRNAs were derived from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were calculated between 11 cirrhotic patients with and without MHE. Next, the biological functions and underlined pathways of DEGs were determined through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Finally, an interactive network between lncRNAs and mRNAs was built, and hub genes were identified, respectively. RESULTS: A total of 64 differentially expressed lncRNAs (dif-lncRNAs) were found between patients with and without MHE, including 30 up- and 34 downregulated genes. 187 differentially expressed mRNAs (dif-mRNAs) were identified, including 84 up- and 103 downregulated genes. Functional enrichment analysis suggested that the regulatory pathways involved in MHE mainly consisted of a series of immune and inflammatory responses. Several hub mRNAs involved in regulatory network were identified, including CCL5, CCR5, CXCR3, CD274, STAT1, CXCR6, and EOMES. In addition, lnc-FAM84B-8 and lnc-SAMD3-1 were found to regulate these above hub genes through building a lncRNA-mRNA network. CONCLUSION: This is the first study to construct the serum lncRNA-mRNA network in MHE, demonstrating the critical role of lncRNAs in regulating inflammatory and immunological profiles in the developing of MHE, suggesting a latent mechanism in this pathophysiological process.


Asunto(s)
Redes Reguladoras de Genes , Encefalopatía Hepática/genética , Cirrosis Hepática/genética , ARN Largo no Codificante/genética , ARN Mensajero/genética , Biología Computacional , Bases de Datos de Ácidos Nucleicos , Regulación hacia Abajo , Perfilación de la Expresión Génica , Ontología de Genes , Encefalopatía Hepática/etiología , Humanos , Cirrosis Hepática/complicaciones , Mapas de Interacción de Proteínas/genética , ARN Largo no Codificante/sangre , ARN Mensajero/sangre , Regulación hacia Arriba
11.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206365

RESUMEN

Acute liver failure (ALF) is associated with deregulated nitric oxide (NO) signaling in the brain, which is one of the key molecular abnormalities leading to the neuropsychiatric disorder called hepatic encephalopathy (HE). This study focuses on the effect of ALF on the relatively unexplored endothelial NOS isoform (eNOS). The cerebral prefrontal cortices of rats with thioacetamide (TAA)-induced ALF showed decreased eNOS expression, which resulted in an overall reduction of NOS activity. ALF also decreased the content of the NOS cofactor, tetrahydro-L-biopterin (BH4), and evoked eNOS uncoupling (reduction of the eNOS dimer/monomer ratio). The addition of the NO precursor L-arginine in the absence of BH4 potentiated ROS accumulation, whereas nonspecific NOS inhibitor L-NAME or EDTA attenuated ROS increase. The ALF-induced decrease of eNOS content and its uncoupling concurred with, and was likely causally related to, both increased brain content of reactive oxidative species (ROS) and decreased cerebral cortical blood flow (CBF) in the same model.


Asunto(s)
Biopterinas/análogos & derivados , Corteza Cerebral/enzimología , Encefalopatía Hepática/enzimología , Fallo Hepático Agudo/enzimología , Óxido Nítrico Sintasa de Tipo III/genética , Animales , Arginina/metabolismo , Biopterinas/análisis , Biopterinas/metabolismo , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/metabolismo , Circulación Cerebrovascular , Regulación de la Expresión Génica , Encefalopatía Hepática/etiología , Encefalopatía Hepática/genética , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/complicaciones , Fallo Hepático Agudo/genética , Masculino , Ratas , Ratas Sprague-Dawley , Tioacetamida/toxicidad
12.
Am J Gastroenterol ; 116(6): 1238-1247, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33852451

RESUMEN

INTRODUCTION: We aimed to define the impact of the genetic background on overt hepatic encephalopathy (HE) in patients with liver cirrhosis by developing a combined clinical-genetic risk score. METHODS: Patients suffering from liver cirrhosis from the outpatient clinics of 4 hospitals (n = 600) were included and followed up for at least 5 years until HE bouts, liver transplant, or death. Patients were genotyped for 60 candidate single nucleotide polymorphisms together with the microsatellite in the promoter region of the gene GLS. RESULTS: Single nucleotide polymorphisms rs601338 (FUT2), rs5743836 (TRL9), rs2562582 (SLC1A3), rs313853 (SLC1A5), and GLS microsatellite did predict independently the incidence and severity of overt HE and were included as genetic score. Competing risk analysis revealed that bilirubin (subhazard ratio [sHR] 1.30 [1.15-1.48], P < 0.001), albumin (sHR 0.90 [0.86-0.93], P < 0.001), genetic score (sHR 1.90 [1.57-2.30], P < 0.001), and previous episodes of overt HE (sHR 2.60 [1.57-4.29], P < 0.001) were independently associated to HE bouts during the follow-up with an internal (C-index 0.83) and external validation (C-index 0.74). Patients in the low-risk group had 5% and 12% risk of HE at 1 (log-rank 92.1; P < 0.001) and 5 (log-rank 124.1; P < 0.001) years, respectively, whereas 36% and 48% in the high-risk group. DISCUSSION: The genetic background influenced overt HE risk and severity. The clinical-genetic HE Risk score, which combined genetic background together with albumin, bilirubin, and previous episodes of overt HE, could be a useful tool to predict overt HE in patients with cirrhosis.


Asunto(s)
Encefalopatía Hepática/genética , Cirrosis Hepática/complicaciones , Medición de Riesgo/métodos , Anciano , Femenino , Genotipo , Encefalopatía Hepática/epidemiología , Humanos , Incidencia , Cirrosis Hepática/etiología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Valor Predictivo de las Pruebas , Índice de Severidad de la Enfermedad , España/epidemiología
13.
Sci Rep ; 11(1): 1907, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479266

RESUMEN

Patients with liver cirrhosis may develop minimal hepatic encephalopathy (MHE) which affects their quality of life and life span. It has been proposed that a shift in peripheral inflammation triggers the appearance of MHE. However, the mechanisms involved in this immune system shift remain unknown. In this work we studied the broad molecular changes involved in the induction of MHE with the goal of identifying (1) altered genes and pathways in peripheral blood cells associated to the appearance of MHE, (2) serum metabolites and cytokines with modified levels in MHE patients and (3) MHE-regulated immune response processes related to changes in specific serum molecules. We adopted a multi-omic approach to profile the transcriptome, metabolome and a panel of cytokines of blood samples taken from cirrhotic patients with or without MHE. Transcriptomic analysis supports the hypothesis of alternations in the Th1/Th2 and Th17 lymphocytes cell populations as major drivers of MHE. Cluster analysis of serum molecules resulted in six groups of chemically similar compounds, suggesting that functional modules operate during the induction of MHE. Finally, the multi-omic integrative analysis suggested a relationship between cytokines CCL20, CX3CL1, CXCL13, IL-15, IL-22 and IL-6 with alteration in chemotaxis, as well as a link between long-chain unsaturated phospholipids and the increased fatty acid transport and prostaglandin production. We found altered immune pathways that may collectively contribute to the mild cognitive impairment phenotype in MHE. Our approach is able to combine extracellular and intracellular information, opening new insights to the understanding of the disease.


Asunto(s)
Citocinas/sangre , Encefalopatía Hepática/genética , Inflamación/genética , Cirrosis Hepática/genética , Vías Biosintéticas/genética , Femenino , Encefalopatía Hepática/sangre , Encefalopatía Hepática/complicaciones , Encefalopatía Hepática/metabolismo , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inflamación/sangre , Inflamación/complicaciones , Inflamación/inmunología , Cirrosis Hepática/sangre , Cirrosis Hepática/complicaciones , Cirrosis Hepática/metabolismo , Linfocitos/metabolismo , Masculino , Metaboloma/genética , Persona de Mediana Edad , Calidad de Vida , Transcriptoma/genética
14.
J Clin Endocrinol Metab ; 106(4): 1084-1090, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33382423

RESUMEN

CONTEXT: Mitchell-Riley syndrome due to RFX6 gene mutations is characterized by neonatal diabetes and protracted diarrhea. The RFX6 gene encodes a transcription factor involved in enteroendocrine cell differentiation required for beta-cell maturation. In contrast to the pathway by which RFX6 mutations leads to diabetes, the mechanisms underlying protracted diarrhea are unknown. OBJECTIVE: To assess whether glucagon-like peptide-1 (GLP-1) was involved in the pathogenesis of Mitchell-Riley syndrome protracted diarrhea. METHODS: Two case report descriptions. in a tertiary pediatric hospital. "Off-label" treatment with liraglutide. We describe 2 children diagnosed with Mitchell-Riley syndrome, presenting neonatal diabetes and protracted diarrhea. Both patients had nearly undetectable GLP-1 plasma levels and absence of GLP-1 immunostaining in distal intestine and rectum. The main outcome was to evaluate whether GLP-1 analogue therapy could improve Mitchell-Riley syndrome protracted diarrhea. RESULTS: "Off-label" liraglutide treatment, licensed for type 2 diabetes treatment in children, was started as rescue therapy for protracted intractable diarrhea resulting in rapid improvement during the course of 12 months. CONCLUSION: Congenital GLP-1 deficiency was identified in patients with Mitchell-Riley syndrome. The favorable response to liraglutide further supports GLP-1 involvement in the pathogenesis of protracted diarrhea and its potential therapeutic use.


Asunto(s)
Diabetes Mellitus/etiología , Diarrea/etiología , Enfermedades de la Vesícula Biliar/etiología , Péptido 1 Similar al Glucagón/deficiencia , Atresia Intestinal/etiología , Niño , Consanguinidad , Diabetes Mellitus/sangre , Diabetes Mellitus/congénito , Diabetes Mellitus/genética , Diarrea/sangre , Diarrea/congénito , Resultado Fatal , Femenino , Enfermedades de la Vesícula Biliar/sangre , Enfermedades de la Vesícula Biliar/congénito , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/fisiología , Encefalopatía Hepática/genética , Encefalopatía Hepática/patología , Humanos , Lactante , Atresia Intestinal/sangre , Mutación Missense , Portugal , Factores de Transcripción del Factor Regulador X/genética
15.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 49(5): 574-580, 2020 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-33210482

RESUMEN

OBJECTIVE: To analyze the clinical phenotype and genetic characteristics of a family with combined oxidative phosphorylation deficiency 1 (COXPD-1). METHODS: The whole exome sequencing was performed in parents of the proband; and the genetic defects were verified by Sanger sequencing technology in the dried blood spot of the proband, the amniotic fluid sample of the little brother of proband, and the peripheral blood of the parents. RESULTS: Whole exome sequencing and Sanger validation showed compound heterozygous mutations of GFM1 gene c.688G>A(p.G230S) and c.1576C>T (p.R526X) in both the proband and her little brother, and the c.1576C>T of GFM1 variant was first reported. The two patients were died in early infancy, and presented with metabolic acidosis, high lactic acid, abnormal liver function, feeding difficulties, microcephaly, development retardation and epilepsy. CONCLUSIONS: GFM1 gene c.688G>A and c.1576C>T compound heterozygous mutations are the cause of this family of COXPD-1.


Asunto(s)
Encefalopatía Hepática , Errores Innatos del Metabolismo , Proteínas Mitocondriales , Factor G de Elongación Peptídica , Resultado Fatal , Femenino , Encefalopatía Hepática/genética , Humanos , Lactante , Masculino , Errores Innatos del Metabolismo/genética , Proteínas Mitocondriales/genética , Mutación , Factor G de Elongación Peptídica/genética , Secuenciación del Exoma
17.
Pediatr Neurol ; 90: 61-65, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391088

RESUMEN

BACKGROUND: Autosomal recessive mutations in the nuclear Twinkle (C10orf2) gene cause a mitochondrial DNA depletion syndrome (MDS) characterized by early onset hepatoencephalopathy. METHODS: We report a severe, early onset encephalopathy and multisystem failure case caused by novel recessive Twinkle gene mutations. Patient clinical, laboratory, and pathological features are reported and Twinkle-associated MDS literature reviewed. RESULTS: Typical presentation includes symptom onset before age six months, failure to thrive, psychomotor regression, epileptic encephalopathy, sensory axonal neuropathy, cholestatic liver dysfunction, and occasionally, renal tubulopathy, movement disorders, and ophthalmoplegia. Death is typical before age four years. CONCLUSIONS: In the differential diagnosis of early onset encephalopathy and multisystem failure, MDS should be considered.


Asunto(s)
ADN Helicasas/genética , ADN Mitocondrial , Encefalopatía Hepática/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Resultado Fatal , Femenino , Encefalopatía Hepática/diagnóstico por imagen , Encefalopatía Hepática/patología , Humanos , Lactante , Recién Nacido , Hígado/patología , Imagen por Resonancia Magnética , Enfermedades Mitocondriales/diagnóstico por imagen , Enfermedades Mitocondriales/patología
18.
J Cell Mol Med ; 22(9): 4056-4067, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29992739

RESUMEN

Acute liver failure (ALF) is associated with high mortality, and a poor understanding of the underlying pathophysiology has resulted in a lack of effective treatments so far. Here, using an amatoxin-induced rhesus monkey model of ALF, we panoramically revealed the cellular and molecular events that lead to the development of ALF. The challenged monkeys with toxins underwent a typical course of ALF including severe hepatic injury, systemic inflammation and eventual death. Adaptive immune was not noticeably disturbed throughout the progress of ALF. A systematic examination of serum factors and cytokines revealed that IL-6 increase was the most rapid and drastic. Interestingly, we found that IL-6 was mainly produced by circulating monocytes. Furthermore, ablation of monocyte-derived IL-6 in mice decreased liver injury and systemic inflammation following chemical injection. Our findings reveal a critical role of circulating monocytes in initiating and accelerating ALF, indicating a potential therapeutic target in clinical treatment for ALF.


Asunto(s)
Amanitinas/toxicidad , Encefalopatía Hepática/inmunología , Interleucina-6/inmunología , Lipopolisacáridos/toxicidad , Fallo Hepático Agudo/inmunología , Monocitos/inmunología , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Citocinas/genética , Citocinas/inmunología , Progresión de la Enfermedad , Expresión Génica , Encefalopatía Hepática/inducido químicamente , Encefalopatía Hepática/genética , Encefalopatía Hepática/patología , Interleucina-6/deficiencia , Interleucina-6/genética , L-Lactato Deshidrogenasa/sangre , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Pruebas de Función Hepática , Macaca mulatta , Ratones , Monocitos/patología
19.
Mol Genet Metab ; 123(1): 28-42, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29331171

RESUMEN

BACKGROUND: Mitochondrial diseases, a group of multi-systemic disorders often characterized by tissue-specific phenotypes, are usually progressive and fatal disorders resulting from defects in oxidative phosphorylation. MTO1 (Mitochondrial tRNA Translation Optimization 1), an evolutionarily conserved protein expressed in high-energy demand tissues has been linked to human early-onset combined oxidative phosphorylation deficiency associated with hypertrophic cardiomyopathy, often referred to as combined oxidative phosphorylation deficiency-10 (COXPD10). MATERIAL AND METHODS: Thirty five cases of MTO1 deficiency were identified and reviewed through international collaboration. The cases of two female siblings, who presented at 1 and 2years of life with seizures, global developmental delay, hypotonia, elevated lactate and complex I and IV deficiency on muscle biopsy but without cardiomyopathy, are presented in detail. RESULTS: For the description of phenotypic features, the denominator varies as the literature was insufficient to allow for complete ascertainment of all data for the 35 cases. An extensive review of all known MTO1 deficiency cases revealed the most common features at presentation to be lactic acidosis (LA) (21/34; 62% cases) and hypertrophic cardiomyopathy (15/34; 44% cases). Eventually lactic acidosis and hypertrophic cardiomyopathy are described in 35/35 (100%) and 27/34 (79%) of patients with MTO1 deficiency, respectively; with global developmental delay/intellectual disability present in 28/29 (97%), feeding difficulties in 17/35 (49%), failure to thrive in 12/35 (34%), seizures in 12/35 (34%), optic atrophy in 11/21 (52%) and ataxia in 7/34 (21%). There are 19 different pathogenic MTO1 variants identified in these 35 cases: one splice-site, 3 frameshift and 15 missense variants. None have bi-allelic variants that completely inactivate MTO1; however, patients where one variant is truncating (i.e. frameshift) while the second one is a missense appear to have a more severe, even fatal, phenotype. These data suggest that complete loss of MTO1 is not viable. A ketogenic diet may have exerted a favourable effect on seizures in 2/5 patients. CONCLUSION: MTO1 deficiency is lethal in some but not all cases, and a genotype-phenotype relation is suggested. Aside from lactic acidosis and cardiomyopathy, developmental delay and other phenotypic features affecting multiple organ systems are often present in these patients, suggesting a broader spectrum than hitherto reported. The diagnosis should be suspected on clinical features and the presence of markers of mitochondrial dysfunction in body fluids, especially low residual complex I, III and IV activity in muscle. Molecular confirmation is required and targeted genomic testing may be the most efficient approach. Although subjective clinical improvement was observed in a small number of patients on therapies such as ketogenic diet and dichloroacetate, no evidence-based effective therapy exists.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Encefalopatía Hepática/genética , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/genética , Adolescente , Biopsia , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/metabolismo , Niño , Preescolar , Femenino , Mutación del Sistema de Lectura , Encefalopatía Hepática/diagnóstico por imagen , Encefalopatía Hepática/fisiopatología , Humanos , Lactante , Recién Nacido , Masculino , Errores Innatos del Metabolismo/diagnóstico por imagen , Errores Innatos del Metabolismo/fisiopatología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Fosforilación Oxidativa , Proteínas de Unión al ARN
20.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 286-295, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29107806

RESUMEN

Hyperammonemia contributes to altered neurotransmission and cognition in patients with hepatic encephalopathy. Hyperammonemia in rats affects differently high- and low-affinity AMPA receptors (AMPARs) in cerebellum. We hypothesized that hyperammonemia would alter differently membrane expression of AMPARs GluA1 and GluA2 subunits by altering its phosphorylation. This work aims were: 1) assess if hyperammonemia alters GluA1 and GluA2 subunits membrane expression in cerebellum and 2) analyze the underlying mechanisms. Hyperammonemia reduces membrane expression of GluA2 and enhances membrane expression of GluA1 in vivo. We show that changes in GluA2 and GluA1 membrane expression in hyperammonemia would be due to enhanced NMDA receptors activation which reduces cGMP levels and phosphodiesterase 2 (PDE2) activity, resulting in increased cAMP levels. This leads to increased protein kinase A (PKA) activity which activates phospholipase C (PLC) and protein kinase C (PKC) thus increasing phosphorylation of GluA2 in Ser880, which reduces GluA2 membrane expression, and phosphorylation of GluA1 in Ser831, which increases GluA1 membrane expression. Blocking NMDA receptors or inhibiting PKA, PLC or PKC normalizes GluA2 and GluA1 phosphorylation and membrane expression in hyperammonemic rats. Altered GluA2 and GluA1 membrane expression would alter signal transduction which may contribute to cognitive and motor alterations in hyperammonemia and hepatic encephalopathy.


Asunto(s)
Membrana Celular/metabolismo , Hiperamonemia/genética , Receptores AMPA/genética , Animales , Membrana Celular/patología , Enfermedad Crónica , Encefalopatía Hepática/genética , Encefalopatía Hepática/metabolismo , Encefalopatía Hepática/patología , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Transducción de Señal/genética , Transmisión Sináptica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA