Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Sci Rep ; 14(1): 8536, 2024 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-38609454

RESUMO

Chronic liver injury induces fibrosis that often proceeds to cirrhosis and hepatocellular carcinoma, indicating that prevention and/or resolution of fibrosis is a promising therapeutic target. Hepatic stellate cells (HSCs) are the major driver of fibrosis by expressing extracellular matrices (ECM). HSCs, in the normal liver, are quiescent and activated by liver injury to become myofibroblasts that proliferate and produce ECM. It has been shown that activated HSCs (aHSCs) become a "quiescent-like" state by removal of liver insults. Therefore, deactivation agents can be a therapeutic drug for advanced liver fibrosis. Using aHSCs prepared from human induced pluripotent stem cells, we found that aHSCs were reverted to a quiescent-like state by a combination of chemical compounds that either inhibit or activate a signaling pathway, Lanifibranor, SB431542, Dorsomorphin, retinoic acid, palmitic acid and Y27632, in vitro. Based on these results, we established a high throughput system to screen agents that induce deactivation and demonstrate that a single chemical compound can induce deactivation.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias Hepáticas , Humanos , Células Estreladas do Fígado , Cirrose Hepática
2.
Biofabrication ; 16(2)2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38447229

RESUMO

Availability of hepatic tissue for the investigation of metabolic processes is severely limited. While primary hepatocytes or animal models are widely used in pharmacological applications, a change in methodology towards more sustainable and ethical assays is highly desirable. Stem cell derived hepatic cells are generally regarded as a viable alternative for the above model systems, if current limitations in functionality and maturation can be overcome. By combining microfluidic organ-on-a-chip technology with individually differentiated, multicellular hepatic tissue fractions, we aim to improve overall functionality of hepatocyte-like cells, as well as evaluate cellular composition and interactions with non-parenchymal cell populations towards the formation of mature liver tissue. Utilizing a multi-omic approach, we show the improved maturation profiles of hepatocyte-like cells maintained in a dynamic microenvironment compared to standard tissue culture setups without continuous perfusion. In order to evaluate the resulting tissue, we employ single cell sequencing to distinguish formed subpopulations and spatial localization. While cellular input was strictly defined based on established differentiation protocols of parenchyma, endothelial and stellate cell fractions, resulting hepatic tissue was shown to comprise a complex mixture of epithelial and non-parenchymal fractions with specific local enrichment of phenotypes along the microchannel. Following this approach, we show the importance of passive, paracrine developmental processes in tissue formation. Using such complex tissue models is a crucial first step to develop stem cell-derivedin vitrosystems that can compare functionally with currently used pharmacological and toxicological applications.


Assuntos
Hepatócitos , Fígado , Animais , Células-Tronco , Diferenciação Celular
3.
Biotechnol Bioeng ; 121(4): 1178-1190, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38184815

RESUMO

Recent advancements in bioengineering have introduced potential alternatives to liver transplantation via the development of self-assembled liver organoids, derived from human-induced pluripotent stem cells (hiPSCs). However, the limited maturity of the tissue makes it challenging to implement this technology on a large scale in clinical settings. In this study, we developed a highly efficient method for generating functional liver organoids from hiPSC-derived carboxypeptidase M liver progenitor cells (CPM+ LPCs), using a microwell structure, and enhanced maturation through direct oxygenation in oxygen-permeable culture plates. We compared the morphology, gene expression profile, and function of the liver organoid with those of cells cultured under conventional conditions using either monolayer or spheroid culture systems. Our results revealed that liver organoids generated using polydimethylsiloxane-based honeycomb microwells significantly exhibited enhanced albumin secretion, hepatic marker expression, and cytochrome P450-mediated metabolism. Additionally, the oxygenated organoids consisted of both hepatocytes and cholangiocytes, which showed increased expression of bile transporter-related genes as well as enhanced bile transport function. Oxygen-permeable polydimethylsiloxane membranes may offer an efficient approach to generating highly mature liver organoids consisting of diverse cell populations.


Assuntos
Células-Tronco Pluripotentes Induzidas , Metaloendopeptidases , Humanos , Oxigênio/metabolismo , Diferenciação Celular , Fígado/metabolismo , Técnicas de Cultura de Células/métodos , Organoides/metabolismo , Dimetilpolisiloxanos , Proteínas Ligadas por GPI
4.
FASEB J ; 38(1): e23359, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38102969

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by severe pruritus and eczematous skin lesions. Although IL-31, a type 2 helper T (Th2)-derived cytokine, is important to the development of pruritus and skin lesions in AD, the blockade of IL-31 signaling does not improve the skin lesions in AD. Oncostatin M (OSM), a member of IL-6 family of cytokines, plays important roles in the regulation of various inflammatory responses through OSM receptor ß subunit (OSMRß), a common receptor subunit for OSM and IL-31. However, the effects of OSM on the pathogenesis of AD remain to be elucidated. When AD model mice were treated with OSM, skin lesions were exacerbated and IL-4 production was increased in the lymph nodes. Next, we investigated the effects of the monoclonal antibody (mAb) against OSMRß on the pathogenesis of AD. Treatment with the anti-OSMRß mAb (7D2) reduced skin severity score in AD model mice. In addition to skin lesions, scratching behavior was decreased by 7D2 mAb with the reduction in the number of OSMRß-positive neurons in the dorsal root ganglia of AD model mice. 7D2 mAb also reduced the serum concentration of IL-4, IL-13, and IgE as well as the gene expressions of IL-4 and IL-13 in the lymph nodes of AD model mice. Blockade of both IL-31 and OSM signaling is suggested to suppress both pruritus and Th2 responses, resulting in the improvement of skin lesions in AD. The anti-OSMRß mAb may be a new therapeutic candidate for the treatment of AD.


Assuntos
Dermatite Atópica , Humanos , Camundongos , Animais , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/metabolismo , Interleucina-13 , Interleucina-4/genética , Pele/metabolismo , Citocinas/metabolismo , Prurido/tratamento farmacológico
5.
Nat Commun ; 14(1): 6304, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37813881

RESUMO

Liver fibrosis results from chronic liver injury triggered by factors such as viral infection, excess alcohol intake, and lipid accumulation. However, the mechanisms underlying liver fibrosis are not fully understood. Here, we demonstrate that the expression of fibroblast growth factor 18 (Fgf18) is elevated in mouse livers following the induction of chronic liver fibrosis models. Deletion of Fgf18 in hepatocytes attenuates liver fibrosis; conversely, overexpression of Fgf18 promotes liver fibrosis. Single-cell RNA sequencing reveals that overexpression of Fgf18 in hepatocytes results in an increase in the number of Lrat+ hepatic stellate cells (HSCs), thereby inducing fibrosis. Mechanistically, FGF18 stimulates the proliferation of HSCs by inducing the expression of Ccnd1. Moreover, the expression of FGF18 is correlated with the expression of profibrotic genes, such as COL1A1 and ACTA2, in human liver biopsy samples. Thus, FGF18 promotes liver fibrosis and could serve as a therapeutic target to treat liver fibrosis.


Assuntos
Células Estreladas do Fígado , Cirrose Hepática , Camundongos , Animais , Humanos , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/patologia , Fígado/metabolismo , Fibrose , Proliferação de Células
6.
Inflamm Regen ; 43(1): 48, 2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37814342

RESUMO

BACKGROUND: The progression of liver fibrosis leads to portal hypertension and liver dysfunction. However, no antifibrotic agents have been approved for cirrhosis to date, making them an unmet medical need. Small extracellular vesicles (sEVs) of mesenchymal stem cells (MSCs) are among these candidate agents. In this study, we investigated the effects of sEVs of MSCs, analyzed their distribution in the liver post-administration, whether their effect was dose-dependent, and whether it was possible to collect a large number of sEVs. METHODS: sEVs expressing tdTomato were generated, and their uptake into constituent liver cells was observed in vitro, as well as their sites of uptake and cells in the liver using a mouse model of liver cirrhosis. The efficiency of sEV collection using tangential flow filtration (TFF) and changes in the therapeutic effects of sEVs in a volume-dependent manner were examined. RESULTS: The sEVs of MSCs accumulated mostly in macrophages in damaged areas of the liver. In addition, the therapeutic effect of sEVs was not necessarily dose-dependent, and it reached a plateau when the dosage exceeded a certain level. Furthermore, although ultracentrifugation was commonly used to collect sEVs for research purposes, we verified that TFF could be used for efficient sEV collection and that their effectiveness is not reduced. CONCLUSION: In this study, we identified some unknown aspects regarding the dynamics, collection, and capacity dependence of sEVs. Our results provide important fundamentals for the development of therapies using sEVs and hold potential implications for the therapeutic applications of sEV-based therapies for liver cirrhosis.

7.
Biotechnol Bioeng ; 120(8): 2345-2356, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37401657

RESUMO

Hepatic stellate cells (HSCs) play a crucial role in liver fibrosis by producing excessive extracellular matrix (ECM) following chronic inflammation. However, studying HSC function has been challenging due to the limited availability of primary human quiescent HSCs (qHSCs) in vitro, and the fact that primary qHSCs quickly activate when cultured on plastic plates. Advances in stem cell technology have allowed for the generation of qHSCs from human induced pluripotent stem cells (hiPSCs) with the potential to provide an unlimited source of cells. However, differentiated quiescent-like HSCs (iqHSCs) also activate spontaneously on conventional plastic plates. In this study, we generated iqHSCs from hiPSCs and developed a culture method to maintain such iqHSCs in a lowly activated state for up to 5 days by optimizing their physical culture microenvironment. We observed that three-dimensional (3D) culture of iqHSCs in soft type 1 collagen hydrogels significantly inhibited their spontaneous activation in vitro while maintaining their ability to convert to activated state. Activation of iqHSC was successfully modeled by stimulating them with the fibrotic cytokine TGFß1. Hence, our culture method can be used to generate HSCs with functions comparable to those in a healthy liver, facilitating the development of accurate in vitro liver models for identifying novel therapeutic agents.


Assuntos
Células Estreladas do Fígado , Células-Tronco Pluripotentes Induzidas , Humanos , Células Estreladas do Fígado/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Diferenciação Celular
8.
Hepatol Res ; 53(7): 661-674, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36866738

RESUMO

AIM: Hepatic zonation is a physiological feature of the liver, known to be key in the regulation of the metabolism of nutrients and xenobiotics and the biotransformation of numerous substances. However, the reproduction of this phenomenon remains challenging in vitro as only part of the processes involved in the orchestration and maintenance of zonation are fully understood. The recent advances in organ-on-chip technologies, which allow for the integration of multicellular 3D tissues in a dynamic microenvironment, could offer solutions for the reproduction of zonation within a single culture vessel. METHODS: An in-depth analysis of zonation-related mechanisms observed during the coculture of human-induced pluripotent stem cell (hiPSC)-derived carboxypeptidase M-positive liver progenitor cells and hiPSC-derived liver sinusoidal endothelial cells within a microfluidic biochip was carried out. RESULTS: Hepatic phenotypes were confirmed in terms of albumin secretion, glycogen storage, CYP450 activity, and expression of specific endothelial markers such as PECAM1, RAB5A, and CD109. Further characterization of the patterns observed in the comparison of the transcription factor motif activities, the transcriptomic signature, and the proteomic profile expressed at the inlet and the outlet of the microfluidic biochip confirmed the presence of zonation-like phenomena within the biochips. In particular, differences related to Wnt/ß-catenin, transforming growth factor-ß, mammalian target of rapamycin, hypoxia-inducible factor-1, and AMP-activated protein kinase signaling, to the metabolism of lipids, and cellular remolding were observed. CONCLUSIONS: The present study shows the interest in combining cocultures of hiPSC-derived cellular models and microfluidic technologies for reproducing in vitro complex mechanisms such as liver zonation and further incites the use of those solutions for accurate reproduction of in vivo situations.

9.
J Pharm Sci ; 112(1): 132-137, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35605686

RESUMO

To develop and assess new dosage forms for the alternative to existing oral medication for peripheral neuropathy, a hydrogel film in the skin patch formation containing tramadol hydrochloride (TRA), a water-soluble drug used as an analgesic, was prepared and evaluated. A hydrogel film composed of 20%(w/w) hydroxypropyl methylcellulose (HPMC) irradiated with electron beams had high transparency and elasticity similar to commercially available wound dressings and soft tissues, suggesting that it is a suitable substrate for TRA. The inclusion of TRA was enabled by immersing the HPMC hydrogel film in TRA aqueous solution. The release and skin permeation of TRA from TRA-containing hydrogel films differed depending on the electron beam dose. Moreover, the analgesic effects in mice were confirmed in a dose-dependent manner. This study demonstrated the usefulness of a hydrogel film containing TRA as a new dosage form alternative to the existing oral medication for peripheral neuropathy.


Assuntos
Neuralgia , Tramadol , Camundongos , Animais , Derivados da Hipromelose , Neuralgia/tratamento farmacológico , Água , Analgésicos , Metilcelulose
10.
Drug Metab Dispos ; 51(2): 174-182, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36379710

RESUMO

Knockout (KO) of mouse Cyp3a genes increases the expression of hepatic CYP2C enzymes, which can metabolize triazolam, a typical substrate of human CYP3A. There is still marked formation of 1'-hydroxytriazolam in Cyp3a-KO (3aKO) mice after triazolam dosing. Here, we generated a new model of humanized CYP3A (hCYP3A) mice with a double-KO background of Cyp3a and Cyp2c genes (2c3aKO), and we examined the metabolic profiles of triazolam in wild-type (WT), 2c3aKO, and hCYP3A/2c3aKO mice in vitro and in vivo In vitro studies using liver microsomes showed that the formation of 1'-hydroxytriazolam in 2c3aKO mice was less than 8% of that in WT mice. The formation rate of 1'-hydroxytriazolam in hCYP3A/2c3aKO mice was eightfold higher than that in 2c3aKO mice. In vivo studies showed that area under the curve (AUC) of 1'-hydroxytriazolam in 2c3aKO mice was less than 3% of that in WT mice. The AUC of 1'-hydroxytriazolam in hCYP3A/2c3aKO mice was sixfold higher than that in 2c3aKO mice. These results showed that formation of 1'-hydroxytriazolam was significantly decreased in 2c3aKO mice. Metabolic functions of human CYP3A enzymes were distinctly found in hCYP3A mice with the 2c3aKO background. Moreover, hCYP3A/2c3aKO mice treated with clobazam showed human CYP3A-mediated formation of desmethylclobazam and prolonged elimination of desmethylclobazam, which is found in poor metabolizers of CYP2C19. The novel hCYP3A mouse model without mouse Cyp2c and Cyp3a genes (hCYP3A/2c3aKO) is expected to be useful to evaluate human CYP3A-mediated metabolism in vivo SIGNIFICANT STATEMENT: Humanized CYP3A (hCYP3A/2c3aKO) mice with a background of double knockout (KO) for mouse Cyp2c and Cyp3a genes were generated. Although CYP2C enzymes played a compensatory role in the metabolism of triazolam to 1'-hydroxytriazolam in the previous hCYP3A/3aKO mice with Cyp2c genes, the novel hCYP3A/2c3aKO mice clearly showed functions of human CYP3A enzymes introduced by chromosome engineering technology.


Assuntos
Triazolam , Humanos , Camundongos , Animais , Triazolam/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Clobazam , Camundongos Knockout , Microssomos Hepáticos/metabolismo
11.
Commun Biol ; 5(1): 85, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35064244

RESUMO

Notch signaling is one of the most common drivers of carcinogenesis in many types of cancers, including hepatocellular carcinoma (HCC); however, it occasionally suppresses tumor progression. Moreover, it is virtually unknown how different sets of Notch ligands and receptors regulate the HCC development. In this study, we demonstrate that the expression of the Notch ligands, Delta-like 4 (Dll4) and Jagged-1 (Jag1), is upregulated during diethylnitrosamine-induced hepatocarcinogenesis. Dll4 is detected in the preneoplastic hepatocytes and HCC cells, but not in the normal hepatocytes, while Jag1 is expressed in the desmin-positive mesenchymal cells. Hepatocyte-specific Dll4 knockout abolishes the Notch1 signaling and suppresses the tumor progression. In contrast, Jag1 deletion induces the ectopic expression of Dll4 in hepatocytes along with the loss of Notch2 signaling, leading to the tumor progression. These results indicate that the two distinct Notch signals, Dll4/Notch1 and Jag1/Notch2, are antagonistic to each other, exerting opposite effects on HCC progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Carcinogênese/metabolismo , Proteína Jagged-1/metabolismo , Neoplasias Hepáticas/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Regulação da Expressão Gênica , Predisposição Genética para Doença , Proteína Jagged-1/genética , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Knockout , Receptor Notch1/genética , Receptor Notch2/genética
12.
Stem Cell Reports ; 16(12): 3050-3063, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34861166

RESUMO

Hepatic stellate cells (HSCs) play a central role in the progression of liver fibrosis by producing extracellular matrices. The development of drugs to suppress liver fibrosis has been hampered by the lack of human quiescent HSCs (qHSCs) and an appropriate in vitro model that faithfully recapitulates HSC activation. In the present study, we developed a culture system to generate qHSC-like cells from human-induced pluripotent stem cells (hiPSCs) that can be converted into activated HSCs in culture. To monitor the activation process, a red fluorescent protein (RFP) gene was inserted in hiPSCs downstream of the activation marker gene actin alpha 2 smooth muscle (ACTA2). Using qHSC-like cells derived from RFP reporter iPSCs, we screened a repurposing chemical library and identified therapeutic candidates that prevent liver fibrosis. Hence, hiPSC-derived qHSC-like cells will be a useful tool to study the mechanism of HSC activation and to identify therapeutic agents.


Assuntos
Técnicas de Cultura de Células , Ciclo Celular , Descoberta de Drogas , Células Estreladas do Fígado/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Animais , Avaliação Pré-Clínica de Medicamentos , Perfilação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Camundongos Endogâmicos C57BL
13.
Differentiation ; 120: 28-35, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34229994

RESUMO

The liver is a complex organ composed of several cell types organized hierarchically. Among these, liver sinusoidal endothelial cells (LSECs) are specialized vascular cells known to interact with hepatocytes and hepatic stellate cells (HSCs), and to be involved in the regulation of important hepatic processes in healthy and pathological situations. Protocols for the differentiation of LSECs from human induced pluripotent stem cells, hiPSCs, have been proposed and in-depth analysis by transcriptomic profiling of those cells has been performed. In the present work, an extended analysis of those cells in terms of proteome and metabolome has been implemented. The proteomic analysis confirmed the expression of important endothelial markers and pathways. Among them, the expression of patterns typical of LSECs such as PECAM1, VWF, LYVE1, STAB1 (endothelial markers), CDH13, CDH5, CLDN5, ICAM1, MCAM-CD146, ICAM2, ESAM (endothelial cytoskeleton), NOSTRIN, NOS3 (Nitric Oxide endothelial ROS), ESM1, ENG, MMRN2, THBS1, ANGPT2 (angiogenesis), CD93, MRC1 (mannose receptor), CLEC14A (C-type lectin), CD40 (antigen), and ERG (transcription factor) was highlighted. Besides, the pathway analysis revealed the enrichment of the endocytosis, Toll-like receptor, Nod-like receptor, Wnt, Apelin, VEGF, cGMP-PCK, and PPAR related signaling pathways. Other important pathways such as vasopressin regulated water reabsorption, fluid shear stress, relaxin signaling, and renin secretion were also highlighted. At confluence, the metabolome profile appeared consistent with quiescent endothelial cell patterns. The integration of both proteome and metabolome datasets revealed a switch from fatty acid synthesis in undifferentiated hiPSCs to a fatty oxidation in LSECs and activation of the pentose phosphate pathway and polyamine metabolism in hiPSCs-derived LSECs. In conclusion, the comparison between the signature of LSECs differentiated following the protocol described in this work, and data found in the literature confirmed the particular relevance of these cells for future in vitro applications.


Assuntos
Diferenciação Celular , Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Metaboloma , Proteoma , Células Cultivadas , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fígado/irrigação sanguínea , Fígado/citologia
14.
Nat Commun ; 12(1): 3390, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099675

RESUMO

In the liver, the bile canaliculi of hepatocytes are connected to intrahepatic bile ducts lined with cholangiocytes, which remove cytotoxic bile from the liver tissue. Although liver organoids have been reported, it is not clear whether the functional connection between hepatocytes and cholangiocytes is recapitulated in those organoids. Here, we report the generation of a hepatobiliary tubular organoid (HBTO) using mouse hepatocyte progenitors and cholangiocytes. Hepatocytes form the bile canalicular network and secrete metabolites into the canaliculi, which are then transported into the biliary tubular structure. Hepatocytes in HBTO acquire and maintain metabolic functions including albumin secretion and cytochrome P450 activities, over the long term. In this study, we establish functional liver tissue incorporating a bile drainage system ex vivo. HBTO enable us to reproduce the transport of hepatocyte metabolites in liver tissue, and to investigate the way in which the two types of epithelial cells establish functional connections.


Assuntos
Ductos Biliares Intra-Hepáticos/citologia , Comunicação Celular/fisiologia , Fígado/citologia , Organoides/fisiologia , Cultura Primária de Células/métodos , Animais , Ductos Biliares Intra-Hepáticos/fisiologia , Diferenciação Celular , Células Cultivadas , Hepatócitos/fisiologia , Fígado/fisiologia , Camundongos , Organoides/citologia , Células-Tronco/fisiologia
15.
Sci Rep ; 11(1): 9530, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33953224

RESUMO

Generation of pancreatic ß cells from pluripotent stem cells is a key technology to develop cell therapy for insulin-dependent diabetes and considerable efforts have been made to produce ß cells. However, due to multiple and lengthy differentiation steps, production of ß cells is often unstable. It is also desirable to eliminate undifferentiated cells to avoid potential risks of tumorigenesis. To isolate ß cell precursors from late stage pancreatic endocrine progenitor (EP) cells derived from iPS cells, we have identified CD82, a member of the tetraspanin family. CD82+ cells at the EP stage differentiated into endocrine cells more efficiently than CD82- EP stage cells. We also show that CD82+ cells in human islets secreted insulin more efficiently than CD82- cells. Furthermore, knockdown of CD82 expression by siRNA or inhibition of CD82 by monoclonal antibodies in NGN3+ cells suppressed the function of ß cells with glucose-stimulated insulin secretion, suggesting that CD82 plays a role in maturation of EP cells to ß cells.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células Secretoras de Insulina/citologia , Proteína Kangai-1/análise , Diferenciação Celular , Linhagem Celular , Separação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Proteína Kangai-1/metabolismo
16.
APL Bioeng ; 5(2): 026104, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34027283

RESUMO

Interactions between the different liver cell types are critical to the maintenance or induction of their function in vitro. In this work, human-induced Pluripotent Stem Cells (hiPSCs)-derived Liver Sinusoidal Endothelial Cells (LSECs) and Hepatocytes-Like Cells (HLCs) were cultured and matured in a microfluidic environment. Both cell populations were differentiated in Petri dishes, detached, and inoculated in microfluidic biochips. In cocultures of both cell types, the tissue has exhibited a higher production of albumin (3.19 vs 5.31 µg/mL/106 cells in monocultures and cocultures) as well as a higher inducibility CYP450 over monocultures of HLCs. Tubular-like structures composed of LSECs and positive for the endothelial marker PECAM1, as well as a tissue more largely expressing Stabilin-2 were detected in cocultures only. In contrast, monocultures exhibited no network and less specific endothelial markers. The transcriptomic analysis did not reveal a marked difference between the profiles of both culture conditions. Nevertheless, the analysis allowed us to highlight different upstream regulators in cocultures (SP1, EBF1, and GATA3) and monocultures (PML, MECP2, and NRF1). In cocultures, the multi-omics dataset after 14 days of maturation in biochips has shown the activation of signaling related to hepatic maturation, angiogenesis, and tissue repair. In this condition, inflammatory signaling was also found to be reduced when compared to monocultures as illustrated by the activation of NFKB and by the detection of several cytokines involved in tissue injury in the latter. Finally, the extracted biological processes were discussed regarding the future development of a new generation of human in vitro hepatic models.

17.
Xenobiotica ; 51(7): 764-770, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34013847

RESUMO

It is important to predict drug-drug interactions via inhibition of intestinal cytochrome P450 3A (CYP3A) which is a determinant of bioavailability of orally administered CYP3A substrates. However, inhibitory effects of macrolide antibiotics on CYP3A-mediated metabolism are not entirely identical between humans and rodents.We investigated the effects of macrolide antibiotics, clarithromycin and erythromycin, on in vitro and in vivo metabolism of triazolam, a CYP3A substrate, in CYP3A-humanised mice generated by using a mouse artificial chromosome vector carrying a human CYP3A gene.Metabolic activities of triazolam were inhibited by macrolide antibiotics in liver and intestine microsomes of CYP3A-humanised mice.The area under the plasma concentration-time curve ratios of 4-hydroxytriazolam to triazolam after oral dosing of triazolam were significantly decreased by multiple administration of macrolide antibiotics. The plasma concentrations ratios of α-hydroxytriazolam and 4-hydroxytriazolam to triazolam in portal blood were significantly decreased by multiple administration of clarithromycin in CYP3A-humanised mice.These results suggest that intestinal CYP3A activity was inhibited by macrolide antibiotics in CYP3A-humanised mice in vitro and in vivo. The plasma concentrations of triazolam and its metabolites in the portal blood of CYP3A-humanised mice would be useful for direct evaluation of intestinal CYP3A-mediated drug-drug interactions.


Assuntos
Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450 , Antibacterianos/farmacologia , Citocromo P-450 CYP3A/genética , Interações Medicamentosas , Humanos , Intestinos , Macrolídeos/farmacologia , Microssomos Hepáticos
18.
NPJ Regen Med ; 6(1): 19, 2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33785758

RESUMO

Mesenchymal stromal cells (MSCs) are used for ameliorating liver fibrosis and aiding liver regeneration after cirrhosis; Here, we analyzed the therapeutic potential of small extracellular vesicles (sEVs) derived from interferon-γ (IFN-γ) pre-conditioned MSCs (γ-sEVs). γ-sEVs effectively induced anti-inflammatory macrophages with high motility and phagocytic abilities in vitro, while not preventing hepatic stellate cell (HSC; the major source of collagen fiber) activation in vitro. The proteome analysis of MSC-derived sEVs revealed anti-inflammatory macrophage inducible proteins (e.g., annexin-A1, lactotransferrin, and aminopeptidase N) upon IFN-γ stimulation. Furthermore, by enabling CX3CR1+ macrophage accumulation in the damaged area, γ-sEVs ameliorated inflammation and fibrosis in the cirrhosis mouse model more effectively than sEVs. Single cell RNA-Seq analysis revealed diverse effects, such as induction of anti-inflammatory macrophages and regulatory T cells, in the cirrhotic liver after γ-sEV administration. Overall, IFN-γ pre-conditioning altered sEVs resulted in efficient tissue repair indicating a new therapeutic strategy.

19.
J Dermatol ; 48(4): 559-563, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33410137

RESUMO

As a novel method of ivermectin (IVM) administration for the treatment of scabies, we devised a whole-body bathing (WBB), in which patients are immersed in a fluid that contains IVM. A multi-institutional trial for elderly patients with scabies was conducted to investigate the efficacy and safety of IVM-WBB. Seven elderly patients with scabies were enrolled and received IVM-WBB up to four times at 1-week interval. The cure for scabies was defined as the absence of mites in two consecutive microscopic or dermoscopic examinations at weekly intervals and the absence of new skin lesions indicative of scabies. Consequently, the cure rate on day 22, the primary end-point, was 71.4%, and all patients had been cured until day 29. Additionally, neither significant adverse events nor clinically problematic abnormal blood test values were obtained. Furthermore, no IVM was detected in the optional plasma (five cases) collected for IVM measurement after bathing. These results suggest that IVM-WBB was effective to treat scabies, causing no serious adverse events and with a very low internal exposure of IVM.


Assuntos
Banhos , Ivermectina , Escabiose , Idoso , Humanos , Ivermectina/uso terapêutico , Escabiose/tratamento farmacológico
20.
Biotechnol Bioeng ; 118(10): 3716-3732, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33404112

RESUMO

Maturation of human-induced pluripotent stem cells (hiPSCs)-derived hepatocytes-like cells (HLCs) toward a complete hepatocyte phenotype remains a challenge as primitiveness patterns are still commonly observed. In this study, we propose a modified differentiation protocol for those cells which includes a prematuration in Petri dishes and a maturation in microfluidic biochip. For the first time, a large range of biomolecular families has been extracted from the same sample to combine transcriptomic, proteomic, and metabolomic analysis. After integration, these datasets revealed specific molecular patterns and highlighted the hepatic regeneration profile in biochips. Overall, biochips exhibited processes of cell proliferation and inflammation (via TGFB1) coupled with anti-fibrotic signaling (via angiotensin 1-7, ATR-2, and MASR). Moreover, cultures in this condition displayed physiological lipid-carbohydrate homeostasis (notably via PPAR, cholesterol metabolism, and bile synthesis) coupled with cell respiration through advanced oxidative phosphorylation (through the overexpression of proteins from the third and fourth complex). The results presented provide an original overview of the complex mechanisms involved in liver regeneration using an advanced in vitro organ-on-chip technology.


Assuntos
Diferenciação Celular , Genômica , Hepatócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Regeneração Hepática , Fígado/metabolismo , Proteômica , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA