Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nat Metab ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867022

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease, encompasses steatosis and metabolic dysfunction-associated steatohepatitis (MASH), leading to cirrhosis and hepatocellular carcinoma. Preclinical MASLD research is mainly performed in rodents; however, the model that best recapitulates human disease is yet to be defined. We conducted a wide-ranging retrospective review (metabolic phenotype, liver histopathology, transcriptome benchmarked against humans) of murine models (mostly male) and ranked them using an unbiased MASLD 'human proximity score' to define their metabolic relevance and ability to induce MASH-fibrosis. Here, we show that Western diets align closely with human MASH; high cholesterol content, extended study duration and/or genetic manipulation of disease-promoting pathways are required to intensify liver damage and accelerate significant (F2+) fibrosis development. Choline-deficient models rapidly induce MASH-fibrosis while showing relatively poor translatability. Our ranking of commonly used MASLD models, based on their proximity to human MASLD, helps with the selection of appropriate in vivo models to accelerate preclinical research.

2.
J Hepatol ; 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38508241

RESUMEN

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) are the key drivers of fibrosis in metabolic dysfunction-associated steatohepatitis (MASH), the fastest growing cause of hepatocellular carcinoma (HCC) worldwide. HSCs are heterogenous, and a senescent subset of HSCs is implicated in hepatic fibrosis and HCC. Administration of anti-uPAR (urokinase-type plasminogen activator receptor) CAR T cells has been shown to deplete senescent HSCs and attenuate fibrosis in murine models. However, the comprehensive features of senescent HSCs in MASH, as well as their cellular ontogeny have not been characterized; hence, we aimed to comprehensively characterize and define the origin of HSCs in human and murine MASH. METHODS: To comprehensively characterize the phenotype and ontogeny of senescent HSCs in human and murine MASH, we integrated senescence-associated beta galactosidase activity with immunostaining, flow cytometry and single-nucleus RNA sequencing (snRNAseq). We integrated the immunohistochemical profile with a senescence score applied to snRNAseq data to characterize senescent HSCs and mapped the evolution of uPAR expression in MASH. RESULTS: Using pseudotime trajectory analysis, we establish that senescent HSCs arise from activated HSCs. While uPAR is expressed in MASH, the magnitude and cell-specificity of its expression evolve with disease stage. In early disease, uPAR is more specific to activated and senescent HSCs, while it is also expressed by myeloid-lineage cells, including Trem2+ macrophages and myeloid-derived suppressor cells, in late disease. Furthermore, we identify novel surface proteins expressed on senescent HSCs in human and murine MASH that could be exploited as therapeutic targets. CONCLUSIONS: These data define features of HSC senescence in human and murine MASH, establishing an important blueprint to target these cells as part of future antifibrotic therapies. IMPACT AND IMPLICATIONS: Hepatic stellate cells (HSCs) are the primary drivers of scarring in chronic liver diseases. As injury develops, a subset of HSCs become senescent; these cells are non-proliferative and pro-inflammatory, thereby contributing to worsening liver injury. Here we show that senescent HSCs are expanded in MASH (metabolic dysfunction-associated steatohepatitis) in humans and mice, and we trace their cellular origin from the activated HSC subset. We further characterize expression of uPAR (urokinase plasminogen activated receptor), a protein that marks senescent HSCs, and report that uPAR is also expressed by activated HSCs in early injury, and in immune cells as liver injury advances. We have integrated high-resolution single-nucleus RNA sequencing with immunostaining and flow cytometry to identify five other novel proteins expressed by senescent HSCs, including mannose receptor CD206, which will facilitate future therapeutic development.

3.
Kidney Int ; 105(3): 540-561, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38159678

RESUMEN

Clinical studies suggest that non-alcoholic steatohepatitis (NASH) is an independent risk factor for chronic kidney disease (CKD), but causality and mechanisms linking these two major diseases are lacking. To assess whether NASH can induce CKD, we have characterized kidney function, histological features, transcriptomic and lipidomic profiles in a well-validated murine NASH model. Mice with NASH progressively developed significant podocyte foot process effacement, proteinuria, glomerulosclerosis, tubular epithelial cell injury, lipid accumulation, and interstitial fibrosis. The progression of kidney fibrosis paralleled the severity of the histologic NASH-activity score. Significantly, we confirmed the causal link between NASH and CKD by orthotopic liver transplantation, which attenuated proteinuria, kidney dysfunction, and fibrosis compared with control sham operated mice. Transcriptomic analysis of mouse kidney cortices revealed differentially expressed genes that were highly enriched in mitochondrial dysfunction, lipid metabolic process, and insulin signaling pathways in NASH-induced CKD. Lipidomic analysis of kidney cortices further revealed that phospholipids and sphingolipids were the most significantly changed lipid species. Notably, we found similar kidney histological changes in human NASH and CKD. Thus, our results confirm a causative role of NASH in the development of CKD, reveal potential pathophysiologic mechanisms of NASH-induced kidney injury, and established a valuable model to study the pathogenesis of NASH-associated CKD. This is an important feature of fatty liver disease that has been largely overlooked but has clinical and prognostic importance.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Insuficiencia Renal Crónica , Humanos , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Insuficiencia Renal Crónica/patología , Fosfolípidos/metabolismo , Proteinuria/patología , Hígado/patología
4.
Sci Transl Med ; 15(677): eadd3949, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36599008

RESUMEN

Advanced hepatic fibrosis, driven by the activation of hepatic stellate cells (HSCs), affects millions worldwide and is the strongest predictor of mortality in nonalcoholic steatohepatitis (NASH); however, there are no approved antifibrotic therapies. To identify antifibrotic drug targets, we integrated progressive transcriptomic and morphological responses that accompany HSC activation in advanced disease using single-nucleus RNA sequencing and tissue clearing in a robust murine NASH model. In advanced fibrosis, we found that an autocrine HSC signaling circuit emerged that was composed of 68 receptor-ligand interactions conserved between murine and human NASH. These predicted interactions were supported by the parallel appearance of markedly increased direct stellate cell-cell contacts in murine NASH. As proof of principle, pharmacological inhibition of one such autocrine interaction, neurotrophic receptor tyrosine kinase 3-neurotrophin 3, inhibited human HSC activation in culture and reversed advanced murine NASH fibrosis. In summary, we uncovered a repertoire of antifibrotic drug targets underlying advanced fibrosis in vivo. The findings suggest a therapeutic paradigm in which stage-specific therapies could yield enhanced antifibrotic efficacy in patients with advanced hepatic fibrosis.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/patología , Células Estrelladas Hepáticas/patología , Comunicación Autocrina , Fibrosis , Cirrosis Hepática/patología , Hígado
5.
Sci Rep ; 12(1): 15661, 2022 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-36123383

RESUMEN

Fatty acid synthase (FASN) is an attractive therapeutic target in non-alcoholic steatohepatitis (NASH) because it drives de novo lipogenesis and mediates pro-inflammatory and fibrogenic signaling. We therefore tested pharmacological inhibition of FASN in human cell culture and in three diet induced mouse models of NASH. Three related FASN inhibitors were used; TVB-3664, TVB-3166 and clinical stage TVB-2640 (denifanstat). In human primary liver microtissues, FASN inhibiton (FASNi) decreased triglyceride (TG) content, consistent with direct anti-steatotic activity. In human hepatic stellate cells, FASNi reduced markers of fibrosis including collagen1α (COL1α1) and α-smooth muscle actin (αSMA). In CD4+ T cells exposed to NASH-related cytokines, FASNi decreased production of Th17 cells, and reduced IL-1ß release in LPS-stimulated PBMCs. In mice with diet induced NASH l, FASNi prevented development of hepatic steatosis and fibrosis, and reduced circulating IL-1ß. In mice with established diet-induced NASH, FASNi reduced NAFLD activity score, fibrosis score, ALT and TG levels. In the CCl4-induced FAT-NASH mouse model, FASN inhibition decreased hepatic fibrosis and fibrosis markers, and development of hepatocellular carcinoma (HCC) tumors by 85%. These results demonstrate that FASN inhibition attenuates inflammatory and fibrotic drivers of NASH by direct inhibition of immune and stellate cells, beyond decreasing fat accumulation in hepatocytes. FASN inhibition therefore provides an opportunity to target three key hallmarks of NASH.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Actinas , Animales , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/prevención & control , Citocinas , Modelos Animales de Enfermedad , Acido Graso Sintasa Tipo I , Ácido Graso Sintasas , Humanos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Nitrilos , Enfermedad del Hígado Graso no Alcohólico/patología , Piperidinas , Triazoles , Triglicéridos
6.
Eur Radiol ; 32(12): 8339-8349, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35727321

RESUMEN

OBJECTIVES: Portal hypertension (PH) is associated with complications such as ascites and esophageal varices and is typically diagnosed through invasive hepatic venous pressure gradient (HVPG) measurement, which is not widely available. In this study, we aim to assess the diagnostic performance of 2D/3D MR elastography (MRE) and shear wave elastography (SWE) measures of liver and spleen stiffness (LS and SS) and spleen volume, to noninvasively diagnose clinically significant portal hypertension (CSPH) using HVPG measurement as the reference. METHODS: In this prospective study, patients with liver disease underwent 2D/3D MRE and SWE of the liver and spleen, as well as HVPG measurement. The correlation between MRE/SWE measures of LS/SS and spleen volume with HVPG was assessed. ROC analysis was used to determine the utility of MRE, SWE, and spleen volume for diagnosing CSPH. RESULTS: Thirty-six patients (M/F 22/14, mean age 55 ± 14 years) were included. Of the evaluated parameters, 3D MRE SS had the strongest correlation with HVPG (r = 0.686, p < 0.001), followed by 2D MRE SS (r = 0.476, p = 0.004). 3D MRE SS displayed the best performance for diagnosis of CSPH (AUC = 0.911) followed by 2D MRE SS (AUC = 0.845) and 3D MRE LS (AUC = 0.804). SWE SS showed poor performance for diagnosis of CSPH (AUC = 0.583) while spleen volume was a fair predictor (AUC = 0.738). 3D MRE SS was significantly superior to SWE LS/SS (p ≤ 0.021) for the diagnosis of CSPH. CONCLUSION: SS measured with 3D MRE outperforms SWE for the diagnosis of CSPH. SS appears to be a useful biomarker for assessing PH severity. These results need further validation. KEY POINTS: • Spleen stiffness measured with 2D and 3D MR elastography correlates significantly with hepatic venous pressure gradient measurement. • Spleen stiffness measured with 3D MR elastography demonstrates excellent performance for the diagnosis of clinically significant portal hypertension (AUC 0.911). • Spleen stiffness measured with 3D MR elastography outperforms liver and spleen stiffness measured with shear wave elastography for diagnosis of clinically significant portal hypertension.


Asunto(s)
Diagnóstico por Imagen de Elasticidad , Hipertensión Portal , Humanos , Adulto , Persona de Mediana Edad , Anciano , Diagnóstico por Imagen de Elasticidad/métodos , Estudios Prospectivos , Cirrosis Hepática/complicaciones , Hipertensión Portal/complicaciones , Hipertensión Portal/diagnóstico por imagen , Hipertensión Portal/patología , Presión Portal , Hígado/patología
7.
Sci Rep ; 11(1): 20827, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675338

RESUMEN

Non-alcoholic steatohepatitis (NASH) is a rising health challenge, with no approved drugs. We used a computational drug repositioning strategy to uncover a novel therapy for NASH, identifying a GABA-B receptor agonist, AZD3355 (Lesogaberan) previously evaluated as a therapy for esophageal reflux. AZD3355's potential efficacy in NASH was tested in human stellate cells, human precision cut liver slices (hPCLS), and in vivo in a well-validated murine model of NASH. In human stellate cells AZD3355 significantly downregulated profibrotic gene and protein expression. Transcriptomic analysis of these responses identified key regulatory nodes impacted by AZD3355, including Myc, as well as MAP and ERK kinases. In PCLS, AZD3355 down-regulated collagen1α1, αSMA and TNF-α mRNAs as well as secreted collagen1α1. In vivo, the drug significantly improved histology, profibrogenic gene expression, and tumor development, which was comparable to activity of obeticholic acid in a robust mouse model of NASH, but awaits further testing to determine its relative efficacy in patients. These data identify a well-tolerated clinical stage asset as a novel candidate therapy for human NASH through its hepatoprotective, anti-inflammatory and antifibrotic mechanisms of action. The approach validates computational methods to identify novel therapies in NASH in uncovering new pathways of disease development that can be rapidly translated into clinical trials.


Asunto(s)
Reposicionamiento de Medicamentos , Agonistas de Receptores GABA-B/uso terapéutico , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Ácidos Fosfínicos/uso terapéutico , Propilaminas/uso terapéutico , Adulto , Anciano , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Agonistas de Receptores GABA-B/farmacología , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Ácidos Fosfínicos/farmacología , Propilaminas/farmacología
8.
J Inherit Metab Dis ; 44(6): 1419-1433, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34564857

RESUMEN

Peroxisomes metabolize a specific subset of fatty acids, which include dicarboxylic fatty acids (DCAs) generated by ω-oxidation. Data obtained in vitro suggest that the peroxisomal transporter ABCD3 (also known as PMP70) mediates the transport of DCAs into the peroxisome, but in vivo evidence to support this role is lacking. In this work, we studied an Abcd3 KO mouse model generated by CRISPR-Cas9 technology using targeted and untargeted metabolomics, histology, immunoblotting, and stable isotope tracing technology. We show that ABCD3 functions in hepatic DCA metabolism and uncover a novel role for this peroxisomal transporter in lipid homeostasis. The Abcd3 KO mouse presents with increased hepatic long-chain DCAs, increased urine medium-chain DCAs, lipodystrophy, enhanced hepatic cholesterol synthesis and decreased hepatic de novo lipogenesis. Moreover, our study suggests that DCAs are metabolized by mitochondrial fatty acid ß-oxidation when ABCD3 is not functional, reflecting the importance of the metabolic compartmentalization and communication between peroxisomes and mitochondria. In summary, this study provides data on the role of the peroxisomal transporter ABCD3 in hepatic lipid homeostasis and DCA metabolism, and the consequences of peroxisomal dysfunction for the liver.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Ácidos Dicarboxílicos/metabolismo , Ácidos Grasos/metabolismo , Homeostasis , Metabolismo de los Lípidos , Transportadoras de Casetes de Unión a ATP/genética , Animales , Femenino , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Oxidación-Reducción , Peroxisomas/metabolismo
9.
JHEP Rep ; 3(3): 100237, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34151243

RESUMEN

BACKGROUND & AIMS: Aramchol is a fatty acid-bile acid conjugate that reduces liver fat content and is being evaluated in a phase III clinical trial for non-alcoholic steatohepatitis (NASH). Aramchol attenuates NASH in mouse models and decreases steatosis by downregulating the fatty acid synthetic enzyme stearoyl CoA desaturase 1 (SCD1) in hepatocytes. Although hepatic stellate cells (HSCs) also store lipids as retinyl esters, the impact of Aramchol in this cell type is unknown. METHODS: We investigated the effects of Aramchol on a human HSC line (LX-2), primary human HSCs (phHSCs), and primary human hepatocytes (phHeps). RESULTS: In LX-2 and phHSCs, 10 µM Aramchol significantly reduced SCD1 mRNA while inducing PPARG (PPARγ) mRNA, with parallel changes in the 2 proteins; ACTA2, COL1A1, ß-PDGFR (bPDGFR) mRNAs were also significantly reduced in LX-2. Secretion of collagen 1 (Col1α1) was inhibited by 10 µM Aramchol. SCD1 knockdown in LX-2 cells phenocopied the effect of Aramchol by reducing fibrogenesis, and addition of Aramchol to these cells did not rescue fibrogenic gene expression. Conversely, in LX-2 overexpressing SCD1, Aramchol no longer suppressed fibrogenic gene expression. The drug also induced genes in LX-2 that promote cholesterol efflux and inhibited ACAT2, which catalyses cholesterol synthesis. In phHeps, Aramchol also reduced SCD1 and increased PPARG mRNA expression. CONCLUSIONS: Aramchol downregulates SCD1 and elevates PPARG in HSCs, reducing COL1A1 and ACTA2 mRNAs and COL1A1 secretion. These data suggest a direct inhibitory effect of Aramchol in HSCs through SCD1 inhibition, as part of a broader impact on both fibrogenic genes as well as mediators of cholesterol homeostasis. These findings illustrate novel mechanisms of Aramchol activity, including potential antifibrotic activity in patients with NASH and fibrosis. LAY SUMMARY: In this study, we have explored the potential activity of Aramchol, a drug currently in clinical trials for fatty liver disease, in blocking fibrosis, or scarring, by hepatic stellate cells, the principal collagen-producing (i.e. fibrogenic) cell type in liver injury. In both isolated human hepatic stellate cells and in a human hepatic stellate cell line, the drug suppresses the key fat-producing enzyme, stearoyl CoA desaturase 1 (SCD1), which leads to reduced expression of genes and proteins associated with hepatic fibrosis, while inducing the protective gene, PPARγ. The drug loses activity when SCD1 is already reduced by gene knockdown, reinforcing the idea that inhibition of SCD1 is a main mode of activity for Aramchol. These findings strengthen the rationale for testing Aramchol in patients with NASH.

10.
Cell Mol Life Sci ; 78(14): 5631-5646, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34110423

RESUMEN

Peroxisomes play an essential role in the ß-oxidation of dicarboxylic acids (DCAs), which are metabolites formed upon ω-oxidation of fatty acids. Genetic evidence linking transporters and enzymes to specific DCA ß-oxidation steps is generally lacking. Moreover, the physiological functions of DCA metabolism remain largely unknown. In this study, we aimed to characterize the DCA ß-oxidation pathway in human cells, and to evaluate the biological role of DCA metabolism using mice deficient in the peroxisomal L-bifunctional protein (Ehhadh KO mice). In vitro experiments using HEK-293 KO cell lines demonstrate that ABCD3 and ACOX1 are essential in DCA ß-oxidation, whereas both the bifunctional proteins (EHHADH and HSD17B4) and the thiolases (ACAA1 and SCPx) have overlapping functions and their contribution may depend on expression level. We also show that medium-chain 3-hydroxydicarboxylic aciduria is a prominent feature of EHHADH deficiency in mice most notably upon inhibition of mitochondrial fatty acid oxidation. Using stable isotope tracing methodology, we confirmed that products of peroxisomal DCA ß-oxidation can be transported to mitochondria for further metabolism. Finally, we show that, in liver, Ehhadh KO mice have increased mRNA and protein expression of cholesterol biosynthesis enzymes with decreased (in females) or similar (in males) rate of cholesterol synthesis. We conclude that EHHADH plays an essential role in the metabolism of medium-chain DCAs and postulate that peroxisomal DCA ß-oxidation is a regulator of hepatic cholesterol biosynthesis.


Asunto(s)
Colesterol/metabolismo , Ácidos Dicarboxílicos/orina , Errores Innatos del Metabolismo Lipídico/patología , Hepatopatías/patología , Mitocondrias/patología , Enzima Bifuncional Peroxisomal/fisiología , Animales , Femenino , Células HEK293 , Homeostasis , Humanos , Errores Innatos del Metabolismo Lipídico/etiología , Hepatopatías/etiología , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo
11.
PLoS One ; 16(1): e0244763, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33395434

RESUMEN

BACKGROUND & AIM: Non-alcoholic steatohepatitis (NASH) is a severe form of non-alcoholic fatty liver disease (NAFLD) that is responsible for a growing fraction of cirrhosis and liver cancer cases worldwide. Changes in the gut microbiome have been implicated in NASH pathogenesis, but the lack of suitable murine models has been a barrier to progress. We have therefore characterized the microbiome in a well-validated murine NASH model to establish its value in modeling human disease. METHODS: The composition of intestinal microbiota was monitored in mice on a 12- or 24-week NASH protocol consisting of high fat, high sugar Western Diet (WD) plus once weekly i.p injection of low-dose CCl4. Additional mice were subjected to WD-only or CCl4-only conditions to assess the independent effect of these variables on the microbiome. RESULTS: There was substantial remodeling of the intestinal microbiome in NASH mice, characterized by declines in both species diversity and bacterial abundance. Based on changes to beta diversity, microbiota from NASH mice clustered separately from controls in principal coordinate analyses. A comparison between WD-only and CCl4-only controls with the NASH model identified WD as the primary driver of early changes to the microbiome, resulting in loss of diversity within the 1st week. A NASH signature emerged progressively at weeks 6 and 12, including, most notably, a reproducible bloom of the Firmicute order Erysipelotrichales. CONCLUSIONS: We have established a valuable model to study the role of gut microbes in NASH, enabling us to identify a new NASH gut microbiome signature.


Asunto(s)
Disbiosis/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Bacterias , Dieta Occidental/efectos adversos , Modelos Animales de Enfermedad , Disbiosis/complicaciones , Heces/microbiología , Fibrosis/complicaciones , Fibrosis/microbiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Variación Genética/genética , Humanos , Inflamación/complicaciones , Cirrosis Hepática/patología , Neoplasias Hepáticas/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/microbiología
12.
Soft Robot ; 8(3): 273-283, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32559391

RESUMEN

The human stomach breaks down and transports food by coordinated radial contractions of the gastric walls. The radial contractions periodically propagate through the stomach and constitute the peristaltic contractions, also called the gastric motility. The force, amplitude, and frequency of peristaltic contractions are relevant to massaging and transporting the food contents in the gastric lumen. However, existing gastric simulators have not faithfully replicated gastric motility. Herein, we report a soft robotic gastric simulator (SoGut) that emulates peristaltic contractions in an anatomically realistic way. SoGut incorporates an array of circular air chambers that generate radial contractions. The design and fabrication of SoGut leverages principles from the soft robotics field, which features compliance and adaptability. We studied the force and amplitude of the contractions when the lumen of SoGut was empty or filled with contents of different viscosity. We examined the contracting force using manometry. SoGut exhibited a similar range of contracting force as the human stomach reported in the literature. Besides, we investigated the amplitude of the contractions through videofluoroscopy where the contraction ratio was derived. The contraction ratio as a function of inflation pressure is found to match the observations of in vivo situations. We demonstrated that SoGut can achieve in vitro peristaltic contractions by coordinating the inflation sequence of multiple air chambers. It exhibited the functions to massage and transport the food contents. SoGut can simulate the physiological motions of the human stomach to advance research of digestion.


Asunto(s)
Robótica , Digestión/fisiología , Humanos , Manometría , Peristaltismo/fisiología , Estómago/fisiología
13.
Soft Robot ; 8(4): 397-415, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32758017

RESUMEN

Soft robotic systems are well suited for developing devices for biomedical applications. A bio-mimicking robotic soft esophagus (RoSE) is developed as an in vitro testing device of endoprosthetic stents for dysphagia management. Endoprosthetic stent placement is an immediate and cost-effective therapy for dysphagia caused by malignant esophageal strictures from esophageal cancer. However, later stage complications, such as stent migration, could weaken the swallow efficacy in the esophagus. The stent radial force (RF) on the esophageal wall is pivotal in avoiding stent migration. Due to limited randomized controlled trials in patients, the stent design and stenting guidelines are still unconstructive. To address the knowledge deficit, we have investigated the capabilities of the RoSE by implanting two stents (stent A and B) of different radial stiffness characteristics, to measure the stent RF and its effect on the stent migration. Also, endoscopic manometry on the RoSE under peristalsis has been performed to study the impact of stenting and stent dysfunctionality on the intrabolus pressure signatures (IBPSs) in the RoSE, and further its effects on the swallowing efficacy. Each implanted stent in the RoSE underwent a set of experiments with various test variables (peristalsis velocity and wavelength, and bolus concentrations). In this study, the conducted tests are representative of the application of RoSE to perform a wide-ranging assessment of the stent behavior. The usability of RoSE has been discussed by comparing the results of stent A and B, for various combinations of the test variables mentioned earlier. The results have demonstrated that the stiffer stent B has a higher RF, whereas stent A maintained its RF at a low profile due to its lesser stiffness. The results have also implicated that a high RF is necessary to minimize the stent migration under prolonged peristaltic contractions in the RoSE. For the manometry experiments, stent A slightly increased the IBPS, but the stiffer stent B significantly decreased the IBPS, especially for the higher concentration boluses. It was found that if a stiffer stent buckles, it can reduce the swallow efficacy and cause recurrent dysphagia. Therefore, RoSE is an innovative soft robotic platform that is capable of testing various endoprosthetic stents, thereby offering a solution to many existing clinical challenges in the area of stent testing.


Asunto(s)
Trastornos de Deglución , Procedimientos Quirúrgicos Robotizados , Robótica , Trastornos de Deglución/etiología , Trastornos de Deglución/terapia , Humanos , Stents/efectos adversos
14.
Sci Rep ; 6: 39342, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27996033

RESUMEN

Autophagy and the unfolded protein response (UPR) both promote activation of hepatic stellate cells (HSC), however the link between the two stimuli remains unclear. Here we have explored the role of X-box binding protein 1 (XBP1), one of three UPR effector pathways and sought to establish the interdependence between autophagy and the UPR during HSC activation. XBP1 induction accompanied both culture-based HSC activation and ER stress induced by tunicamycin. Ectopic overexpression of XBP1 induced collagen 1-alpha expression in HSCs, which was inhibited by knockdown of ATG7, a critical autophagy mediator. Genome-wide transcriptomic profiling indicated an upregulation of collagen synthesis pathways, but not of the transforming growth factor (TGF)-b pathway, a canonical fibrogenic driver, suggesting that XBP1 activates a specific subset of fibrogenesis pathways independent of TGF-ß1. XBP1 target gene signatures were significantly induced in rodent liver fibrosis models (n = 3-5) and in human samples of non-alcoholic fatty liver disease (NAFLD) (n = 72-135). Thus, XBP1-mediated UPR contributes to fibrogenic HSC activation and is functionally linked to cellular autophagy.


Asunto(s)
Autofagia/fisiología , Células Estrelladas Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Respuesta de Proteína Desplegada/fisiología , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/genética , Línea Celular , Colágeno Tipo I/biosíntesis , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HEK293 , Humanos , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño , Tunicamicina/efectos adversos , Proteína 1 de Unión a la X-Box/genética
15.
Global Health ; 12(1): 75, 2016 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-27884162

RESUMEN

BACKGROUND: Donors commonly fund innovative interventions to improve health in the hope that governments of low and middle-income countries will scale-up those that are shown to be effective. Yet innovations can be slow to be adopted by country governments and implemented at scale. Our study explores this problem by identifying key contextual factors influencing scale-up of maternal and newborn health innovations in three low-income settings: Ethiopia, the six states of northeast Nigeria and Uttar Pradesh state in India. METHODS: We conducted 150 semi-structured interviews in 2012/13 with stakeholders from government, development partner agencies, externally funded implementers including civil society organisations, academic institutions and professional associations to understand scale-up of innovations to improve the health of mothers and newborns these study settings. We analysed interview data with the aid of a common analytic framework to enable cross-country comparison, with Nvivo to code themes. RESULTS: We found that multiple contextual factors enabled and undermined attempts to catalyse scale-up of donor-funded maternal and newborn health innovations. Factors influencing government decisions to accept innovations at scale included: how health policy decisions are made; prioritising and funding maternal and newborn health; and development partner harmonisation. Factors influencing the implementation of innovations at scale included: health systems capacity in the three settings; and security in northeast Nigeria. Contextual factors influencing beneficiary communities' uptake of innovations at scale included: sociocultural contexts; and access to healthcare. CONCLUSIONS: We conclude that context is critical: externally funded implementers need to assess and adapt for contexts if they are to successfully position an innovation for scale-up.


Asunto(s)
Difusión de Innovaciones , Política de Salud , Salud del Lactante/estadística & datos numéricos , Servicios de Salud Materna/organización & administración , Salud Materna/estadística & datos numéricos , Etiopía , Femenino , Humanos , India , Recién Nacido , Nigeria , Embarazo , Investigación Cualitativa
16.
PLoS One ; 10(6): e0131399, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26114725

RESUMEN

The dihydropyridine receptor (DHPR) ß1a subunit is essential for skeletal muscle excitation-contraction coupling, but the structural organization of ß1a as part of the macromolecular DHPR-ryanodine receptor type I (RyR1) complex is still debatable. We used fluorescence resonance energy transfer (FRET) to probe proximity relationships within the ß1a subunit in cultured skeletal myotubes lacking or expressing RyR1. The fluorescein biarsenical reagent FlAsH was used as the FRET acceptor, which exhibits fluorescence upon binding to specific tetracysteine motifs, and enhanced cyan fluorescent protein (CFP) was used as the FRET donor. Ten ß1a reporter constructs were generated by inserting the CCPGCC FlAsH binding motif into five positions probing the five domains of ß1a with either carboxyl or amino terminal fused CFP. FRET efficiency was largest when CCPGCC was positioned next to CFP, and significant intramolecular FRET was observed for all constructs suggesting that in situ the ß1a subunit has a relatively compact conformation in which the carboxyl and amino termini are not extended. Comparison of the FRET efficiency in wild type to that in dyspedic (lacking RyR1) myotubes revealed that in only one construct (H458 CCPGCC ß1a -CFP) FRET efficiency was specifically altered by the presence of RyR1. The present study reveals that the C-terminal of the ß1a subunit changes conformation in the presence of RyR1 consistent with an interaction between the C-terminal of ß1a and RyR1 in resting myotubes.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Secuencias de Aminoácidos , Animales , Canales de Calcio Tipo L/genética , Ratones , Ratones Mutantes , Fibras Musculares Esqueléticas , Canal Liberador de Calcio Receptor de Rianodina/genética
17.
Soc Sci Med ; 121: 30-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25306407

RESUMEN

Donors and other development partners commonly introduce innovative practices and technologies to improve health in low and middle income countries. Yet many innovations that are effective in improving health and survival are slow to be translated into policy and implemented at scale. Understanding the factors influencing scale-up is important. We conducted a qualitative study involving 150 semi-structured interviews with government, development partners, civil society organisations and externally funded implementers, professional associations and academic institutions in 2012/13 to explore scale-up of innovative interventions targeting mothers and newborns in Ethiopia, the Indian state of Uttar Pradesh and the six states of northeast Nigeria, which are settings with high burdens of maternal and neonatal mortality. Interviews were analysed using a common analytic framework developed for cross-country comparison and themes were coded using Nvivo. We found that programme implementers across the three settings require multiple steps to catalyse scale-up. Advocating for government to adopt and finance health innovations requires: designing scalable innovations; embedding scale-up in programme design and allocating time and resources; building implementer capacity to catalyse scale-up; adopting effective approaches to advocacy; presenting strong evidence to support government decision making; involving government in programme design; invoking policy champions and networks; strengthening harmonisation among external programmes; aligning innovations with health systems and priorities. Other steps include: supporting government to develop policies and programmes and strengthening health systems and staff; promoting community uptake by involving media, community leaders, mobilisation teams and role models. We conclude that scale-up has no magic bullet solution - implementers must embrace multiple activities, and require substantial support from donors and governments in doing so.


Asunto(s)
Servicios de Salud del Niño/organización & administración , Implementación de Plan de Salud/métodos , Política de Salud , Servicios de Salud Materna/organización & administración , Desarrollo de Programa , Países en Desarrollo , Etiopía , Femenino , Humanos , India , Lactante , Recién Nacido , Nigeria , Investigación Cualitativa
18.
J Gen Virol ; 95(Pt 2): 363-372, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23997183

RESUMEN

Non-structural 5A protein (NS5A) has emerged as an important pharmacological target for hepatitis C virus (HCV). However, little is known about the conformation of NS5A intracellularly or how NS5A inhibitors achieve the picomolar (pM) inhibition of virus replication. Here, we have presented two structurally related small molecules, one that potently inhibits HCV replication and selects for resistance in NS5A, and another that is inactive. Resistance to this antiviral was greater in genotype 1a than in genotype 1b replicons and mapped to domain 1 of NS5A. Using a novel cell-based assay that measures the intracellular proximity of fluorescent tags covalently attached to NS5A, we showed that only the active antiviral specifically disrupted the close proximity of inter- and intramolecular positions of NS5A. The active antiviral, termed compound 1, caused a repositioning of both the N and C termini of NS5A, including disruption of the close approximation of the N termini of two different NS5A molecules in a multimolecular complex. These data provide the first study of how antivirals that select resistance in domain 1 of NS5A alter the cellular conformation of NS5A. This class of antiviral disrupts the close proximity of the N termini of domain 1 in a NS5A complex but also alters the conformation of domain 3, and leads to large aggregates of NS5A. Current models predict that a multicomponent cocktail of antivirals is needed to treat HCV infection, so a mechanistic understanding of what each component does to the viral machinery will be important.


Asunto(s)
Antivirales/metabolismo , Hepacivirus/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Línea Celular , Farmacorresistencia Viral , Hepatocitos/virología , Humanos , Conformación Proteica/efectos de los fármacos , Selección Genética , Replicación Viral/efectos de los fármacos
19.
PLoS One ; 8(11): e81820, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244748

RESUMEN

Apart from few early biophysical studies, the relative thermal instability of HbE has been only shown by clinical investigations. We have compared in vitro thermal stability of HbE with HbA2 and HbA using optical spectroscopy. From absorption measurements in the soret region, synchronous fluorescence spectroscopy and dynamic light scattering experiments, we have found thermal stability of the three hemoglobin variants following the order HbE11.0 in all the three variants. Under oxidative stress conditions in presence of hydrogen peroxide, HbE has been found to be more vulnerable to aggregation compared to HbA and HbA2. Taken together, these studies have shown thermal and oxidative instability of HbE and points towards the role of HbE in the upregulation of redox regulators and chaperone proteins in erythrocyte proteome of patients suffering from HbEbeta thalassemia.


Asunto(s)
Hemoglobina A/química , Hemoglobina A/metabolismo , Hemoglobina E/química , Hemoglobina E/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Oxidación-Reducción , Pliegue de Proteína , Estabilidad Proteica
20.
Vector Borne Zoonotic Dis ; 13(8): 590-600, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23930976

RESUMEN

Many arboviral proteins are phosphorylated in infected mammalian cells, but it is unknown if the same phosphorylation events occur when insects are similarly infected. One of the mammalian kinases responsible for phosphorylation, protein kinase G (PKG), has been implicated in the behavior of multiple nonvector insects, but is unstudied in mosquitoes. PKG from Aedes aegypti was cloned, and phosphorylation of specific viral sites was monitored by mass spectrometry from biochemical and cell culture experiments. PKG from Aedes mosquitoes is able to phosphorylate dengue nonstructural protein 5 (NS5) at specific sites in cell culture and cell-free systems and autophosphorylates its own regulatory domain in a cell-free system. Injecting Aedes aegypti and Anopheles gambiae mosquitoes with a pharmacological PKG activator resulted in increased Aedes wing activity during periods of their natural diurnal/crepuscular activity and increased Anopheles nocturnal locomotor/flight activity. Thus, perturbation of the PKG signaling pathway in mosquitoes alters flight behavior. The demonstrated effect of PKG alterations is consistent with a viral PKG substrate triggering increased PKG activity. This increased PKG activity could be the mechanism by which dengue virus increases flight behavior and possibly facilitates transmission. Whether or not PKG is part of the mechanism by which dengue increases flight behavior, this report is the first to show PKG can modulate behavior in hematophagous disease vectors.


Asunto(s)
Aedes/enzimología , Anopheles/enzimología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Virus del Dengue/metabolismo , Proteínas no Estructurales Virales/metabolismo , Aedes/efectos de los fármacos , Aedes/fisiología , Aedes/virología , Secuencia de Aminoácidos , Animales , Anopheles/efectos de los fármacos , Anopheles/fisiología , Anopheles/virología , Conducta Animal , Técnicas de Cultivo de Célula , Sistema Libre de Células , Femenino , Vuelo Animal , Humanos , Espectrometría de Masas , Datos de Secuencia Molecular , Fosforilación , Alineación de Secuencia , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA