Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Nat Nanotechnol ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956320

RESUMO

Neuromodulation technologies are crucial for investigating neuronal connectivity and brain function. Magnetic neuromodulation offers wireless and remote deep brain stimulations that are lacking in optogenetic- and wired-electrode-based tools. However, due to the limited understanding of working principles and poorly designed magnetic operating systems, earlier magnetic approaches have yet to be utilized. Furthermore, despite its importance in neuroscience research, cell-type-specific magnetic neuromodulation has remained elusive. Here we present a nanomaterials-based magnetogenetic toolbox, in conjunction with Cre-loxP technology, to selectively activate genetically encoded Piezo1 ion channels in targeted neuronal populations via torque generated by the nanomagnetic actuators in vitro and in vivo. We demonstrate this cell-type-targeting magnetic approach for remote and spatiotemporal precise control of deep brain neural activity in multiple behavioural models, such as bidirectional feeding control, long-term neuromodulation for weight control in obese mice and wireless modulation of social behaviours in multiple mice in the same physical space. Our study demonstrates the potential of cell-type-specific magnetogenetics as an effective and reliable research tool for life sciences, especially in wireless, long-term and freely behaving animals.

2.
BMB Rep ; 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38919019

RESUMO

The utilization of multi-omics research has gained popularity in clinical investigations. However, effectively managing and merging extensive and diverse datasets presents a challenge due to its intricacy. This research introduces a Multi-Omics Analysis Sandbox Toolkit, an online platform designed to facilitate the exploration, integration, and visualization of datasets ranging from single-omics to multi-omics. This platform establishes connections between clinical data and omics information, allowing for versatile analysis and storage of both single and multi-omics data. Additionally, users can repeatedly utilize and exchange their findings within the platform. This toolkit offers diverse alternatives for data selection and gene set analysis. It also presents visualization outputs, potential candidates, and annotations. Furthermore, this platform empowers users to collaborate by sharing their datasets, analyses, and conclusions with others, thus enhancing its utility as a collaborative research tool. This Multi-Omics Analysis Sandbox Toolkit stands as a valuable asset in comprehensively grasping the influence of diverse factors in diseases and pinpointing potential biomarkers.

3.
Biochem Biophys Res Commun ; 696: 149512, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38224664

RESUMO

Epithelial cell adhesion molecules (EpCAMs) have been identified as surface markers of proliferating ductal cells, which are referred to as liver progenitor cells (LPCs), during liver regeneration and correspond to malignancies. These cells can differentiate into hepatocytes and biliary epithelial cells (BECs) in vitro. EpCAM-positive LPCs are involved in liver regeneration following severe liver injury; however, the in vivo function of EpCAMs in the regenerating liver remains unclear. In the present study, we used a zebrafish model of LPC-driven liver regeneration to elucidate the function of EpCAMs in the regenerating liver in vivo. Proliferating ductal cells were observed after severe hepatocyte loss in the zebrafish model. Analyses of the liver size as well as hepatocyte and BEC markers revealed successful conversion of LPCs to hepatocytes and BECs in epcam mutants. Notably, epcam mutants exhibited severe defects in intrahepatic duct maturation and bile acid secretion in regenerating hepatocytes, suggesting that epcam plays a critical role in intrahepatic duct reconstruction during LPC-driven liver regeneration. Our findings provide insights into human diseases involving non-parenchymal cells, such as primary biliary cholangitis, by highlighting the regulatory effect of epcam on intrahepatic duct reconstruction.


Assuntos
Colangite , Peixe-Zebra , Animais , Humanos , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/metabolismo , Fígado/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Hepatócitos/metabolismo , Células Epiteliais/metabolismo , Colangite/patologia , Regeneração Hepática
4.
Cell Stem Cell ; 30(12): 1640-1657.e8, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38029740

RESUMO

The liver is known for its remarkable regenerative ability through proliferation of hepatocytes. Yet, during chronic injury or severe hepatocyte death, proliferation of hepatocytes is exhausted. To overcome this hurdle, we propose vascular-endothelial-growth-factor A (VEGFA) as a therapeutic means to accelerate biliary epithelial-cell (BEC)-to-hepatocyte conversion. Investigation in zebrafish establishes that blocking VEGF receptors abrogates BEC-driven liver repair, while VEGFA overexpression promotes it. Delivery of VEGFA via nonintegrative and safe nucleoside-modified mRNA encapsulated into lipid nanoparticles (mRNA-LNPs) in acutely or chronically injured mouse livers induces robust BEC-to-hepatocyte conversion and elimination of steatosis and fibrosis. In human and murine diseased livers, we further identified VEGFA-receptor KDR-expressing BECs associated with KDR-expressing cell-derived hepatocytes. This work defines KDR-expressing cells, most likely being BECs, as facultative progenitors. This study reveals unexpected therapeutic benefits of VEGFA delivered via nucleoside-modified mRNA-LNP, whose safety is widely validated with COVID-19 vaccines, for harnessing BEC-driven repair to potentially treat liver diseases.


Assuntos
Hepatopatias , Peixe-Zebra , Animais , Camundongos , Humanos , RNA Mensageiro/genética , Vacinas contra COVID-19 , Nucleosídeos , Hepatócitos , Fígado , Células Epiteliais , Hepatopatias/patologia , Fibrose , Regeneração Hepática , Fator A de Crescimento do Endotélio Vascular/genética
5.
Sci Rep ; 13(1): 18312, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880271

RESUMO

Despite the robust regenerative capacity of the liver, prolonged and severe liver damage impairs liver regeneration, leading to liver failure. Since the liver co-opts the differentiation of liver progenitor cells (LPCs) into hepatocytes to restore functional hepatocytes, augmenting LPC-mediated liver regeneration may be beneficial to patients with chronic liver diseases. However, the molecular mechanisms underlying LPC-to-hepatocyte differentiation have remained largely unknown. Using the zebrafish model of LPC-mediated liver regeneration, Tg(fabp10a:pt-ß-catenin), we present that peroxisome proliferator-activated receptor-alpha (PPARα) activation augments LPC-to-hepatocyte differentiation. We found that treating Tg(fabp10a:pt-ß-catenin) larvae with GW7647, a potent PPARα agonist, enhanced the expression of hepatocyte markers and simultaneously reduced the expression of biliary epithelial cell (BEC)/LPC markers in the regenerating livers, indicating enhanced LPC-to-hepatocyte differentiation. Mechanistically, PPARα activation augments the differentiation by suppressing YAP signaling. The differentiation phenotypes resulting from GW7647 treatment were rescued by expressing a constitutively active form of Yap1. Moreover, we found that suppression of YAP signaling was sufficient to promote LPC-to-hepatocyte differentiation. Treating Tg(fabp10a:pt-ß-catenin) larvae with the TEAD inhibitor K-975, which suppresses YAP signaling, phenocopied the effect of GW7647 on LPC differentiation. Altogether, our findings provide insights into augmenting LPC-mediated liver regeneration as a regenerative therapy for chronic liver diseases.


Assuntos
Hepatopatias , PPAR alfa , Proteínas de Sinalização YAP , Peixe-Zebra , Animais , beta Catenina/metabolismo , Proliferação de Células , Hepatócitos/metabolismo , Fígado/metabolismo , Hepatopatias/metabolismo , Regeneração Hepática/fisiologia , PPAR alfa/metabolismo , Células-Tronco/metabolismo , Peixe-Zebra/genética
6.
J Hepatol ; 79(6): 1385-1395, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37572794

RESUMO

BACKGROUND & AIMS: Biliary atresia (BA) is poorly understood and leads to liver transplantation (LT), with the requirement for and associated risks of lifelong immunosuppression, in most children. We performed a genome-wide association study (GWAS) to determine the genetic basis of BA. METHODS: We performed a GWAS in 811 European BA cases treated with LT in US, Canadian and UK centers, and 4,654 genetically matched controls. Whole-genome sequencing of 100 cases evaluated synthetic association with rare variants. Functional studies included whole liver transcriptome analysis of 64 BA cases and perturbations in experimental models. RESULTS: A GWAS of common single nucleotide polymorphisms (SNPs), i.e. allele frequencies >1%, identified intronic SNPs rs6446628 in AFAP1 with genome-wide significance (p = 3.93E-8) and rs34599046 in TUSC3 at sub-threshold genome-wide significance (p = 1.34E-7), both supported by credible peaks of neighboring SNPs. Like other previously reported BA-associated genes, AFAP1 and TUSC3 are ciliogenesis and planar polarity effectors (CPLANE). In gene-set-based GWAS, BA was associated with 6,005 SNPs in 102 CPLANE genes (p = 5.84E-15). Compared with non-CPLANE genes, more CPLANE genes harbored rare variants (allele frequency <1%) that were assigned Human Phenotype Ontology terms related to hepatobiliary anomalies by predictive algorithms, 87% vs. 40%, p <0.0001. Rare variants were present in multiple genes distinct from those with BA-associated common variants in most BA cases. AFAP1 and TUSC3 knockdown blocked ciliogenesis in mouse tracheal cells. Inhibition of ciliogenesis caused biliary dysgenesis in zebrafish. AFAP1 and TUSC3 were expressed in fetal liver organoids, as well as fetal and BA livers, but not in normal or disease-control livers. Integrative analysis of BA-associated variants and liver transcripts revealed abnormal vasculogenesis and epithelial tube formation, explaining portal vein anomalies that co-exist with BA. CONCLUSIONS: BA is associated with polygenic susceptibility in CPLANE genes. Rare variants contribute to polygenic risk in vulnerable pathways via unique genes. IMPACT AND IMPLICATIONS: Liver transplantation is needed to cure most children born with biliary atresia, a poorly understood rare disease. Transplant immunosuppression increases the likelihood of life-threatening infections and cancers. To improve care by preventing this disease and its progression to transplantation, we examined its genetic basis. We find that this disease is associated with both common and rare mutations in highly specialized genes which maintain normal communication and movement of cells, and their organization into bile ducts and blood vessels during early development of the human embryo. Because defects in these genes also cause other birth defects, our findings could lead to preventive strategies to lower the incidence of biliary atresia and potentially other birth defects.


Assuntos
Atresia Biliar , Criança , Animais , Camundongos , Humanos , Atresia Biliar/genética , Estudo de Associação Genômica Ampla , Predisposição Genética para Doença , Peixe-Zebra/genética , Canadá
7.
bioRxiv ; 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-37131823

RESUMO

The liver is known for its remarkable regenerative ability through proliferation of hepatocytes. Yet, during chronic injury or severe hepatocyte death, proliferation of hepatocytes is exhausted. To overcome this hurdle, we propose vascular-endothelial-growth-factor A (VEGFA) as a therapeutic means to accelerate biliary epithelial cell (BEC)-to-hepatocyte conversion. Investigation in zebrafish establishes that blocking VEGF receptors abrogates BEC-driven liver repair, while VEGFA overexpression promotes it. Delivery of VEGFA via non-integrative and safe nucleoside-modified mRNA encapsulated into lipid-nanoparticles (mRNA-LNP) in acutely or chronically injured mouse livers induces robust BEC-to-hepatocyte conversion and reversion of steatosis and fibrosis. In human and murine diseased livers, we further identified VEGFA-receptor KDR-expressing BECs associated with KDR-expressing cell-derived hepatocytes. This defines KDR-expressing cells, most likely being BECs, as facultative progenitors. This study reveals novel therapeutic benefits of VEGFA delivered via nucleoside-modified mRNA-LNP, whose safety is widely validated with COVID-19 vaccines, for harnessing BEC-driven repair to potentially treat liver diseases. Highlights: Complementary mouse and zebrafish models of liver injury demonstrate the therapeutic impact of VEGFA-KDR axis activation to harness BEC-driven liver regeneration.VEGFA mRNA LNPs restore two key features of the chronic liver disease in humans such as steatosis and fibrosis.Identification in human cirrhotic ESLD livers of KDR-expressing BECs adjacent to clusters of KDR+ hepatocytes suggesting their BEC origin.KDR-expressing BECs may represent facultative adult progenitor cells, a unique BEC population that has yet been uncovered.

8.
Clin Hypertens ; 29(1): 13, 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-37122032

RESUMO

The prevalence of hypertension (HT) among young adults aged 18 to 39 years is estimated to be 3.7% to 8.6% worldwide. Although the prevalence of HT in young adults is lower than that of the overall population, those with HT are at substantially increased risk of cardiovascular events compared to those without HT. HT in young adults should be taken with even more caution as longer exposure to higher blood pressure leads to a higher lifetime risk of HT-mediated organ damage. However, young patients with HT show low awareness of HT compared to older patients. Also, they are more prone to show low treatment adherence despite the good efficacy of the treatment. Other risk factors that hinder HT control among young adults include alcohol intake, smoking, low physical activity, emotional stress, job stress, metabolic syndrome, and obesity. This review aimed to illustrate the suboptimal control status of the young hypertensive population and to propose strategies for improvement.

9.
Semin Liver Dis ; 43(1): 13-23, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36764306

RESUMO

The liver field has been debating for decades the contribution of the plasticity of the two epithelial compartments in the liver, hepatocytes and biliary epithelial cells (BECs), to derive each other as a repair mechanism. The hepatobiliary plasticity has been first observed in diseased human livers by the presence of biphenotypic cells expressing hepatocyte and BEC markers within bile ducts and regenerative nodules or budding from strings of proliferative BECs in septa. These observations are not surprising as hepatocytes and BECs derive from a common fetal progenitor, the hepatoblast, and, as such, they are expected to compensate for each other's loss in adults. To investigate the cell origin of regenerated cell compartments and associated molecular mechanisms, numerous murine and zebrafish models with ability to trace cell fates have been extensively developed. This short review summarizes the clinical and preclinical studies illustrating the hepatobiliary plasticity and its potential therapeutic application.


Assuntos
Fígado , Peixe-Zebra , Animais , Camundongos , Humanos , Hepatócitos , Células Epiteliais
10.
Hepatology ; 77(4): 1198-1210, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36626626

RESUMO

BACKGROUND AND AIMS: Injury to biliary epithelial cells (BECs) lining the hepatic bile ducts leads to cholestatic liver diseases. Upon severe biliary damage, hepatocytes can convert to BECs, thereby contributing to liver recovery. Given a potential of augmenting this hepatocyte-to-BEC conversion as a therapeutic option for cholestatic liver diseases, it will be important to thoroughly understand the cellular and molecular mechanisms of the conversion process. APPROACH AND RESULTS: Towards this aim, we have established a zebrafish model for hepatocyte-to-BEC conversion by employing Tg(fabp10a:CFP-NTR) zebrafish with a temporal inhibition of Notch signaling during regeneration. Cre/loxP-mediated permanent and H2B-mCherry-mediated short-term lineage tracing revealed that in the model, all BECs originate from hepatocytes. During the conversion, BEC markers are sequentially induced in the order of Sox9b, Yap/Taz, Notch activity/ epcam , and Alcama/ krt18 ; the expression of the hepatocyte marker Bhmt disappears between the Sox9b and Yap/Taz induction. Importantly, live time-lapse imaging unambiguously revealed transdifferentiation of hepatocytes into BECs: hepatocytes convert to BECs without transitioning through a proliferative intermediate state. In addition, using compounds and transgenic and mutant lines that modulate Notch and Yap signaling, we found that both Notch and Yap signaling are required for the conversion even in Notch- and Yap-overactivating settings. CONCLUSIONS: Hepatocyte-to-BEC conversion occurs through transdifferentiation independently of proliferation, and Notch and Yap signaling control the process in parallel with a mutually positive interaction. The new zebrafish model will further contribute to a thorough understanding of the mechanisms of the conversion process.


Assuntos
Colestase , Hepatopatias , Animais , Peixe-Zebra , Transdiferenciação Celular/fisiologia , Hepatócitos/metabolismo , Fígado , Células Epiteliais , Colestase/metabolismo , Hepatopatias/metabolismo , Proliferação de Células , Regeneração Hepática/fisiologia
11.
Hepatology ; 77(3): 789-801, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35829917

RESUMO

BACKGROUND AND AIMS: Hepatocytes were the first cell type for which oscillations of cytoplasmic calcium levels in response to hormones were described. Since then, investigation of calcium dynamics in liver explants and culture has greatly increased our understanding of calcium signaling. A bottleneck, however, exists in observing calcium dynamics in a noninvasive manner because of the optical inaccessibility of the mammalian liver. Here, we aimed to take advantage of the transparency of the zebrafish larvae to image hepatocyte calcium dynamics in vivo at cellular resolution. APPROACH AND RESULTS: We developed a transgenic model expressing a calcium sensor, GCaMP6s, specifically in zebrafish hepatocytes. Using this, we provide a quantitative assessment of intracellular calcium dynamics during multiple contexts, including growth, feeding, ethanol-induced stress, and cell ablation. Specifically, we show that synchronized calcium oscillations are present in vivo , which are lost upon starvation. Starvation induces lipid accumulation in the liver. Feeding recommences calcium waves in the liver, but in a spatially restricted manner, as well as resolves starvation-induced hepatic steatosis. By using a genetically encoded scavenger for calcium, we show that dampening of calcium signaling accelerates the accumulation of starvation-related lipid droplets in the liver. Furthermore, ethanol treatment, as well as cell ablation, induces calcium flux, but with different dynamics. The former causes asynchronous calcium oscillations, whereas the latter leads to a single calcium spike. CONCLUSIONS: We demonstrate the presence of oscillations, waves, and spikes in vivo . Calcium waves are present in response to nutrition and negatively regulate starvation-induced accumulation of lipid droplets.


Assuntos
Inanição , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Cálcio/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Etanol/farmacologia , Sinalização do Cálcio , Inanição/metabolismo , Mamíferos/metabolismo
12.
Sensors (Basel) ; 22(19)2022 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-36236744

RESUMO

Object detection is an essential function for mobile robots, allowing them to carry out missions efficiently. In recent years, various deep learning models based on convolutional neural networks have achieved good performance in object detection. However, in cases in which robots have to carry out missions in a particular environment, utilizing a model that has been trained without considering the environment in which robots must conduct their tasks degrades their object detection performance, leading to failed missions. This poor model accuracy occurs because of the class imbalance problem, in which the occurrence frequencies of the object classes in the training dataset are significantly different. In this study, we propose a systematic solution that can solve the class imbalance problem by training multiple object detection models and using these models effectively for robots that move through various environments to carry out missions. Moreover, we show through experiments that the proposed multi-model-based object detection framework with environment-context awareness can effectively overcome the class imbalance problem. As a result of the experiment, CPU usage decreased by 45.49% and latency decreased by more than 60%, while object detection accuracy increased by 6.6% on average.


Assuntos
Robótica , Redes Neurais de Computação
13.
Hepatology ; 75(2): 322-337, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34435364

RESUMO

BACKGROUND AND AIMS: In patients with acute liver failure (ALF) who suffer from massive hepatocyte loss, liver progenitor cells (LPCs) take over key hepatocyte functions, which ultimately determines survival. This study investigated how the expression of hepatocyte nuclear factor 4α (HNF4α), its regulators, and targets in LPCs determines clinical outcome of patients with ALF. APPROACH AND RESULTS: Clinicopathological associations were scrutinized in 19 patients with ALF (9 recovered and 10 receiving liver transplantation). Regulatory mechanisms between follistatin, activin, HNF4α, and coagulation factor expression in LPC were investigated in vitro and in metronidazole-treated zebrafish. A prospective clinical study followed up 186 patients with cirrhosis for 80 months to observe the relevance of follistatin levels in prevalence and mortality of acute-on-chronic liver failure. Recovered patients with ALF robustly express HNF4α in either LPCs or remaining hepatocytes. As in hepatocytes, HNF4α controls the expression of coagulation factors by binding to their promoters in LPC. HNF4α expression in LPCs requires the forkhead box protein H1-Sma and Mad homolog 2/3/4 transcription factor complex, which is promoted by the TGF-ß superfamily member activin. Activin signaling in LPCs is negatively regulated by follistatin, a hepatocyte-derived hormone controlled by insulin and glucagon. In contrast to patients requiring liver transplantation, recovered patients demonstrate a normal activin/follistatin ratio, robust abundance of the activin effectors phosphorylated Sma and Mad homolog 2 and HNF4α in LPCs, leading to significantly improved coagulation function. A follow-up study indicated that serum follistatin levels could predict the incidence and mortality of acute-on-chronic liver failure. CONCLUSIONS: These results highlight a crucial role of the follistatin-controlled activin-HNF4α-coagulation axis in determining the clinical outcome of massive hepatocyte loss-induced ALF. The effects of insulin and glucagon on follistatin suggest a key role of the systemic metabolic state in ALF.


Assuntos
Ativinas/genética , Folistatina/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Falência Hepática Aguda/metabolismo , Ativinas/metabolismo , Insuficiência Hepática Crônica Agudizada/sangue , Adulto , Idoso , Animais , Coagulação Sanguínea , Linhagem Celular , Fator V/genética , Feminino , Folistatina/sangue , Seguimentos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Hepatócitos/metabolismo , Humanos , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/patologia , Falência Hepática Aguda/cirurgia , Regeneração Hepática , Transplante de Fígado , Masculino , Metronidazol , Camundongos , Pessoa de Meia-Idade , Prognóstico , Regiões Promotoras Genéticas , Estudos Prospectivos , Protrombina/genética , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteína Smad4/genética , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta1/genética , Peixe-Zebra
14.
Chemosphere ; 292: 133336, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34922955

RESUMO

Total organic carbon (TOC), total nitrogen (TN), and total phosphorous (TP) are the most common indicators of water quality. The analytical processes of the indicators require oxidation of any type of C, N, and P to carbon dioxide, nitrate, and phosphate as final products. Persulfate is the recommended oxidant for these transformations. In this study, co-oxidation was suggested for the simultaneous analysis of TOC-TN-TP. A single oxidizing reactor using persulfate was proposed instead of three individual systems. The system oxidizes complex organic chemicals to carbon dioxide, nitrate, and phosphate. However, the residual persulfate after oxidation interferes with the spectrophotometric analysis of nitrate and phosphate. Therefore, in the proposed system, the complete transformation of persulfate is a key factor for simultaneous analysis. In this system, ultraviolet irradiation for 30 min under alkaline conditions converted residual persulfate to sulfate. The complete transformation eliminated persulfate interference in nitrate and phosphate detection. In the proposed system with a single oxidation reactor, TOC, TN, and TP were oxidized and analyzed simultaneously within 15% of the analytical error range compared to the standard test method.


Assuntos
Nitrogênio , Poluentes Químicos da Água , Oxirredução , Fósforo , Sulfatos
15.
Elife ; 102021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34609282

RESUMO

Expansion of biliary epithelial cells (BECs) during ductular reaction (DR) is observed in liver diseases including cystic fibrosis (CF), and associated with inflammation and fibrosis, albeit without complete understanding of underlying mechanism. Using two different genetic mouse knockouts of ß-catenin, one with ß-catenin loss is hepatocytes and BECs (KO1), and another with loss in only hepatocytes (KO2), we demonstrate disparate long-term repair after an initial injury by 2-week choline-deficient ethionine-supplemented diet. KO2 show gradual liver repopulation with BEC-derived ß-catenin-positive hepatocytes and resolution of injury. KO1 showed persistent loss of ß-catenin, NF-κB activation in BECs, progressive DR and fibrosis, reminiscent of CF histology. We identify interactions of ß-catenin, NFκB, and CF transmembranous conductance regulator (CFTR) in BECs. Loss of CFTR or ß-catenin led to NF-κB activation, DR, and inflammation. Thus, we report a novel ß-catenin-NFκB-CFTR interactome in BECs, and its disruption may contribute to hepatic pathology of CF.


The liver has an incredible capacity to repair itself or 'regenerate' ­ that is, it has the ability to replace damaged tissue with new tissue. In order to do this, the organ relies on hepatocytes (the cells that form the liver) and bile duct cells (the cells that form the biliary ducts) dividing and transforming into each other to repair and replace damaged tissue, in case the insult is dire. During long-lasting or chronic liver injury, bile duct cells undergo a process called 'ductular reaction', which causes the cells to multiply and produce proteins that stimulate inflammation, and can lead to liver scarring (fibrosis). Ductular reaction is a hallmark of severe liver disease, and different diseases exhibit ductular reactions with distinct features. For example, in cystic fibrosis, a unique type of ductular reaction occurs at late stages, accompanied by both inflammation and fibrosis. Despite the role that ductular reaction plays in liver disease, it is not well understood how it works at the molecular level. Hu et al. set out to investigate how a protein called ß-catenin ­ which can cause many types of cells to proliferate ­ is involved in ductular reaction. They used three types of mice for their experiments: wild-type mice, which were not genetically modified; and two strains of genetically modified mice. One of these mutant mice did not produce ß-catenin in biliary duct cells, while the other lacked ß-catenin both in biliary duct cells and in hepatocytes. After a short liver injury ­ which Hu et al. caused by feeding the mice a specific diet ­ the wild-type mice were able to regenerate and repair the liver without exhibiting any ductular reaction. The mutant mice that lacked ß-catenin in hepatocytes showed a temporary ductular reaction, and ultimately repaired their livers by turning bile duct cells into hepatocytes. On the other hand, the mutant mice lacking ß-catenin in both hepatocytes and bile duct cells displayed sustained ductular reactions, inflammation and fibrosis, which looked like that seen in patients with liver disease associated to cystic fibrosis. Further probing showed that ß-catenin interacts with a protein called CTFR, which is involved in cystic fibrosis. When bile duct cells lack either of these proteins, another protein called NF-B gets activated, which causes the ductular reaction, leading to inflammation and fibrosis. The findings of Hu et al. shed light on the role of ß-catenin in ductular reaction. Further, the results show a previously unknown interaction between ß-catenin, CTFR and NF-B, which could lead to better treatments for cystic fibrosis in the future.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose/genética , Inflamação/genética , NF-kappa B/genética , beta Catenina/genética , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Fibrose/imunologia , Inflamação/imunologia , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , beta Catenina/metabolismo
16.
Sci Total Environ ; 789: 147961, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34052499

RESUMO

In-situ chemical oxidation (ISCO) is commonly practiced to degrade organic pollutants in various fields. However, ISCO is deteriorated the oxidation efficiency due to the non-selective and self-decomposition of reagents. Therefore, in-situ generation of oxidants is being proposed to compensate for the demerits of conventional ISCO. In this study, the aim is to suggest a novel in-situ generation system using the combination of electrochemical oxidation (EO) and pyrite oxidation. It is hypothesized that EO system can generate the oxygen species, which can activate the pyrite surface to produce more oxidants. We evaluated three systems (1) EO system (2) pyrite oxidation system (3) combined system using sulfanilamide as a common antibiotic. The EO system degraded completely sulfanilamide and generated 150 µM of H2O2 and 8 mg/L of DO even at 10 mA. In other words, EO system can directly oxidize the sulfanilamide and produce oxygen species. The pyrite system produced 204 and 24 µM of hydroxyl radicals at pH 3 under oxic and anoxic conditions, respectively, and 118 and 20 µM at pH 7. Pyrite oxidation can generate more reactive species in the presence of oxygen. The combined system enhanced the oxidation-rate constant to 1.5 times (from 0.2561 to 0.3502 h-1). The additional supply of oxygen showed a higher oxidation rate to 1.5 and 1.3 times higher than single EO or pyrite oxidation, respectively. As a result, the co-presence of pyrite and oxygen shows a synergistic effect on the oxidation of the organic pollutant. Our results suggest that electrochemical generation of the oxygen species in the presence of pyrite is a promising technique to oxidize organic pollutants in groundwater.


Assuntos
Peróxido de Hidrogênio , Sulfetos , Radical Hidroxila , Oxirredução , Espécies Reativas de Oxigênio
17.
Hepatology ; 74(1): 397-410, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33314176

RESUMO

BACKGROUND AND AIMS: Following mild liver injury, pre-existing hepatocytes replicate. However, if hepatocyte proliferation is compromised, such as in chronic liver diseases, biliary epithelial cells (BECs) contribute to hepatocytes through liver progenitor cells (LPCs), thereby restoring hepatic mass and function. Recently, augmenting innate BEC-driven liver regeneration has garnered attention as an alternative to liver transplantation, the only reliable treatment for patients with end-stage liver diseases. Despite this attention, the molecular basis of BEC-driven liver regeneration remains poorly understood. APPROACH AND RESULTS: By performing a chemical screen with the zebrafish hepatocyte ablation model, in which BECs robustly contribute to hepatocytes, we identified farnesoid X receptor (FXR) agonists as inhibitors of BEC-driven liver regeneration. Here we show that FXR activation blocks the process through the FXR-PTEN (phosphatase and tensin homolog)-PI3K (phosphoinositide 3-kinase)-AKT-mTOR (mammalian target of rapamycin) axis. We found that FXR activation blocked LPC-to-hepatocyte differentiation, but not BEC-to-LPC dedifferentiation. FXR activation also suppressed LPC proliferation and increased its death. These defects were rescued by suppressing PTEN activity with its chemical inhibitor and ptena/b mutants, indicating PTEN as a critical downstream mediator of FXR signaling in BEC-driven liver regeneration. Consistent with the role of PTEN in inhibiting the PI3K-AKT-mTOR pathway, FXR activation reduced the expression of pS6, a marker of mTORC1 activation, in LPCs of regenerating livers. Importantly, suppressing PI3K and mTORC1 activities with their chemical inhibitors blocked BEC-driven liver regeneration, as did FXR activation. CONCLUSIONS: FXR activation impairs BEC-driven liver regeneration by enhancing PTEN activity; the PI3K-AKT-mTOR pathway controls the regeneration process. Given the clinical trials and use of FXR agonists for multiple liver diseases due to their beneficial effects on steatosis and fibrosis, the detrimental effects of FXR activation on LPCs suggest a rather personalized use of the agonists in the clinic.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Regeneração Hepática/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/agonistas , Células-Tronco/efeitos dos fármacos , Animais , Animais Geneticamente Modificados , Sistema Biliar/citologia , Proliferação de Células , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Fígado/efeitos dos fármacos , Fígado/fisiologia , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Células-Tronco/fisiologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
18.
Hepatology ; 73(4): 1494-1508, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32602149

RESUMO

BACKGROUND AND AIMS: The liver is a highly regenerative organ, but its regenerative capacity is compromised in severe liver injury settings. In chronic liver diseases, the number of liver progenitor cells (LPCs) correlates proportionally to disease severity, implying that their inefficient differentiation into hepatocytes exacerbates the disease. Moreover, LPCs secrete proinflammatory cytokines; thus, their prolonged presence worsens inflammation and induces fibrosis. Promoting LPC-to-hepatocyte differentiation in patients with advanced liver disease, for whom liver transplantation is currently the only therapeutic option, may be a feasible clinical approach because such promotion generates more functional hepatocytes and concomitantly reduces inflammation and fibrosis. APPROACH AND RESULTS: Here, using zebrafish models of LPC-mediated liver regeneration, we present a proof of principle of such therapeutics by demonstrating a role for the epidermal growth factor receptor (EGFR) signaling pathway in differentiation of LPCs into hepatocytes. We found that suppression of EGFR signaling promoted LPC-to-hepatocyte differentiation through the mitogen-activated ERK kinase (MEK)-extracellular signal-regulated kinase (ERK)-sex-determining region Y-box 9 (SOX9) cascade. Pharmacological inhibition of EGFR or MEK/ERK promoted LPC-to-hepatocyte differentiation as well as genetic suppression of the EGFR-ERK-SOX9 axis. Moreover, Sox9b overexpression in LPCs blocked their differentiation into hepatocytes. In the zebrafish liver injury model, both hepatocytes and biliary epithelial cells contributed to LPCs. EGFR inhibition promoted the differentiation of LPCs regardless of their origin. Notably, short-term treatment with EGFR inhibitors resulted in better liver recovery over the long term. CONCLUSIONS: The EGFR-ERK-SOX9 axis suppresses LPC-to-hepatocyte differentiation during LPC-mediated liver regeneration. We suggest EGFR inhibitors as a proregenerative therapeutic drug for patients with advanced liver disease.


Assuntos
Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regeneração Hepática/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Butadienos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Hepatócitos/citologia , Nitrilas/farmacologia , Quinazolinas/farmacologia , Células-Tronco/citologia , Tirfostinas/farmacologia
19.
J Hazard Mater ; 401: 123746, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33113729

RESUMO

Total organic carbon (TOC) has been suggested and utilized as an index of organic matter in aqueous phases. The overall performance of TOC is highly dependent on the method of oxidation of organic matter to carbon dioxide, such as high-temperature combustion (HTC) and wet chemical oxidation (WCO). HTC requires more energy and maintenance cost, it is a major barrier to the field application. In contrast, WCO is more suitable for the application of on-line monitoring systems due to requiring lower energy and easy maintenance. However, WCO shows lower oxidation than HTC, thus, oxidation performance should be improved for the application to the field. In this study, a dual radical system (DRS), including sulfate and hydroxyl radicals, was proposed to enhance oxidation ability. The DRS uses alkaline pH and persulfate to generate sulfate radicals, which have been used to activate hydroxyl radicals and oxidize organic matter. The oxidation mechanism for the DRS has been verified using model chemicals with different reaction rate constants. The applicability of the DRS has been confirmed using authentic wastewater with a high concentration of chloride. In this study, the DRS showed similar performance compared to the HTC within 10 % error range. The DRS shows similar oxidation performance with HTC even at a high concentration of chloride. DRS did not show interference by the presence of chloride up to 30,000 mg/L of chloride. Results of this study indicate that the DRS can enhance overall oxidation performance compared to the conventional WCO system.

20.
Front Physiol ; 11: 538701, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33192543

RESUMO

BACKGROUND/AIMS: Infectious and genetic factors are invoked, respectively in isolated biliary atresia (BA), or syndromic BA, with major extrahepatic anomalies. However, isolated BA is also associated with minor extrahepatic gut and cardiovascular anomalies and multiple susceptibility genes, suggesting common origins. METHODS: We investigated novel susceptibility genes with genome-wide association, targeted sequencing and tissue staining in BA requiring liver transplantation, independent of BA subtype. Candidate gene effects on morphogenesis, developmental pathways, and ciliogenesis, which regulates left-right patterning were investigated with zebrafish knockdown and mouse knockout models, mouse airway cell cultures, and liver transcriptome analysis. RESULTS: Single nucleotide polymorphisms in Mannosidase-1-α-2 (MAN1A2) were significantly associated with BA and with other polymorphisms known to affect MAN1A2 expression but were not differentially enriched in either BA subtype. In zebrafish embryos, man1a2 knockdown caused poor biliary network formation, ciliary dysgenesis in Kupffer's vesicle, cardiac and liver heterotaxy, and dysregulated egfra and other developmental genes. Suboptimal man1a2 knockdown synergized with suboptimal EGFR signaling or suboptimal knockdown of the EGFR pathway gene, adenosine-ribosylation-factor-6, which had minimal effects individually, to reproduce biliary defects but not heterotaxy. In cultured mouse airway epithelium, Man1a2 knockdown arrested ciliary development and motility. Man1a2 -/- mice, which experience respiratory failure, also demonstrated portal and bile ductular inflammation. Human BA liver and Man1a2 -/- liver exhibited reduced Man1a2 expression and dysregulated ciliary genes, known to cause multisystem human laterality defects. CONCLUSION: BA requiring transplantation associates with sequence variants in MAN1A2. man1a2 regulates laterality, in addition to hepatobiliary morphogenesis, by regulating ciliogenesis in zebrafish and mice, providing a novel developmental basis for multisystem defects in BA.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...