Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 620
Filter
1.
Biochem J ; 481(18): 1173-1186, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39171361

ABSTRACT

Pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor that plays a key role in drug metabolism. Recently, PXR was found to attenuate the development of liver cancer by suppressing epithelial-mesenchymal transition (EMT) in liver cancer cells in a mouse model of two-stage chemical carcinogenesis. To elucidate the role of PXR in the EMT of liver cancer cells, we focused on its role in hepatic stellate cells (HSCs), which are components of the tumor microenvironment in hepatocellular carcinoma (HCC). Human HSC-derived LX-2 cells stably expressed destabilization domain (DD)-fused human PXR (hPXR-LX2 cells). Human HCC-derived HepG2 cells were transfected with the EMT marker VIM promoter-regulated reporter plasmid and co-cultured with hPXR-LX2 cells or treated with hPXR-LX2-derived conditioned medium (CM). Co-culture or CM treatment increased reporter activity in HepG2 cells. This induction was attenuated upon PXR activation in hPXR-LX2 cells by treatment with the DD-stabilizing chemical Shield-1 and the human PXR ligand rifampicin. PXR activation in hPXR-LX2 cells exhibited inhibition of TGF-ß1-induced transdifferentiation, supported by observations of morphological changes and protein or mRNA levels of the transdifferentiation markers COL1A1 and FN1. PXR activation in hPXR-LX2 cells also attenuated the mRNA levels of the key transdifferentiation factor, POSTN. Treatment of hPXR-LX2 cells with recombinant POSTN restored the PXR-mediated suppression of transdifferentiation. Reporter assays with the POSTN promoter showed that PXR inhibited the NF-κB-mediated transcription of POSTN. Consequently, PXR activation in HSCs is expected to inhibit transdifferentiation by down-regulating POSTN expression, thereby suppressing EMT of liver cancer cells.


Subject(s)
Cell Adhesion Molecules , Cell Transdifferentiation , Down-Regulation , Hepatic Stellate Cells , Pregnane X Receptor , Humans , Pregnane X Receptor/metabolism , Pregnane X Receptor/genetics , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Cell Transdifferentiation/drug effects , Hep G2 Cells , Down-Regulation/drug effects , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Epithelial-Mesenchymal Transition/drug effects , Periostin
2.
FASEB J ; 38(15): e23848, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39092889

ABSTRACT

Glucocorticoid use may cause elevated intraocular pressure, leading to the development of glucocorticoid-induced glaucoma (GIG). However, the mechanism of GIG development remains incompletely understood. In this study, we subjected primary human trabecular meshwork cells (TMCs) and mice to dexamethasone treatment to mimic glucocorticoid exposure. The myofibroblast transdifferentiation of TMCs was observed in cellular and mouse models, as well as in human trabecular mesh specimens. This was demonstrated by the cytoskeletal reorganization, alterations in cell morphology, heightened transdifferentiation markers, increased extracellular matrix deposition, and cellular dysfunction. Knockdown of Rho guanine nucleotide exchange factor 26 (ARHGEF26) expression ameliorated dexamethasone-induced changes in cell morphology and upregulation of myofibroblast markers, reversed dysfunction and extracellular matrix deposition in TMCs, and prevented the development of dexamethasone-induced intraocular hypertension. And, this process may be related to the TGF-ß pathway. In conclusion, glucocorticoids induced the myofibroblast transdifferentiation in TMCs, which played a crucial role in the pathogenesis of GIG. Inhibition of ARHGEF26 expression protected TMCs by reversing myofibroblast transdifferentiation. This study demonstrated the potential of reversing the myofibroblast transdifferentiation of TMCs as a new target for treating GIG.


Subject(s)
Cell Transdifferentiation , Dexamethasone , Glaucoma , Myofibroblasts , Rho Guanine Nucleotide Exchange Factors , Trabecular Meshwork , Dexamethasone/pharmacology , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Trabecular Meshwork/cytology , Cell Transdifferentiation/drug effects , Animals , Humans , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/cytology , Mice , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Glaucoma/pathology , Glaucoma/metabolism , Cells, Cultured , Glucocorticoids/pharmacology , Mice, Inbred C57BL , Male
3.
Front Immunol ; 15: 1412022, 2024.
Article in English | MEDLINE | ID: mdl-38881898

ABSTRACT

Abdominal aortic aneurysm (AAA) is a degenerative disease characterized by local abnormal dilation of the aorta accompanied by vascular smooth muscle cell (VSMC) dysfunction and chronic inflammation. VSMC dedifferentiation, transdifferentiation, and increased expression of matrix metalloproteinases (MMPs) are essential causes of AAA formation. Previous studies from us and others have shown that Anemoside B4 (AB4), a saponin from Pulsatilla chinensis, has anti-inflammatory, anti-tumor, and regulatory effects on VSMC dedifferentiation. The current study aimed to investigate whether AB4 inhibits AAA development and its underlying mechanisms. By using an Ang II induced AAA model in vivo and cholesterol loading mediated VSMC to macrophage transdifferentiation model in vitro, our study demonstrated that AB4 could attenuate AAA pathogenesis, prevent VSMC dedifferentiation and transdifferentiation to macrophage-like cells, decrease vascular inflammation, and suppress MMP expression and activity. Furthermore, KLF4 overexpression attenuated the effects of AB4 on VSMC to macrophage-like cell transition and VSMC inflammation in vitro. In conclusion, AB4 protects against AAA formation in mice by inhibiting KLF4 mediated VSMC transdifferentiation and inflammation. Our study provides the first proof of concept of using AB4 for AAA management.


Subject(s)
Aortic Aneurysm, Abdominal , Cell Transdifferentiation , Inflammation , Kruppel-Like Factor 4 , Myocytes, Smooth Muscle , Saponins , Animals , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Aortic Aneurysm, Abdominal/chemically induced , Cell Transdifferentiation/drug effects , Kruppel-Like Factor 4/metabolism , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Inflammation/metabolism , Saponins/pharmacology , Disease Models, Animal , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/drug effects , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Angiotensin II/pharmacology , Humans
4.
Aging (Albany NY) ; 16(11): 9876-9898, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38843385

ABSTRACT

Estrogen is thought to have a role in slowing down aging and protecting cardiovascular and cognitive function. However, high doses of estrogen are still positively associated with autoimmune diseases and tumors with systemic inflammation. First, we administered exogenous estrogen to female mice for three consecutive months and found that the aorta of mice on estrogen develops inflammatory manifestations similar to Takayasu arteritis (TAK). Then, in vitro estrogen intervention was performed on mouse aortic vascular smooth muscle cells (MOVAS cells). Stimulated by high concentrations of estradiol, MOVAS cells showed decreased expression of contractile phenotypic markers and increased expression of macrophage-like phenotypic markers. This shift was blocked by tamoxifen and Krüppel-like factor 4 (KLF4) inhibitors and enhanced by Von Hippel-Lindau (VHL)/hypoxia-inducible factor-1α (HIF-1α) interaction inhibitors. It suggests that estrogen-targeted regulation of the VHL/HIF-1α/KLF4 axis induces phenotypic transformation of vascular smooth muscle cells (VSMC). In addition, estrogen-regulated phenotypic conversion of VSMC to macrophages is a key mechanism of estrogen-induced vascular inflammation, which justifies the risk of clinical use of estrogen replacement therapy.


Subject(s)
Estrogens , Hypoxia-Inducible Factor 1, alpha Subunit , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Macrophages , Muscle, Smooth, Vascular , Von Hippel-Lindau Tumor Suppressor Protein , Animals , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Macrophages/metabolism , Macrophages/drug effects , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/drug effects , Female , Estrogens/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Cell Transdifferentiation/drug effects , Phenotype , Aorta/pathology , Aorta/drug effects , Inflammation/metabolism
5.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727276

ABSTRACT

In mammals, hearing loss is irreversible due to the lack of the regenerative capacity of the auditory epithelium. However, stem/progenitor cells in mammalian cochleae may be a therapeutic target for hearing regeneration. The ubiquitin proteasome system plays an important role in cochlear development and maintenance. In this study, we investigated the role of ubiquitin C-terminal hydrolase L1 (UCHL1) in the process of the transdifferentiation of auditory supporting cells (SCs) into hair cells (HCs). The expression of UCHL1 gradually decreased as HCs developed and was restricted to inner pillar cells and third-row Deiters' cells between P2 and P7, suggesting that UCHL1-expressing cells are similar to the cells with Lgr5-positive progenitors. UCHL1 expression was decreased even under conditions in which supernumerary HCs were generated with a γ-secretase inhibitor and Wnt agonist. Moreover, the inhibition of UCHL1 by LDN-57444 led to an increase in HC numbers. Mechanistically, LDN-57444 increased mTOR complex 1 activity and allowed SCs to transdifferentiate into HCs. The suppression of UCHL1 induces the transdifferentiation of auditory SCs and progenitors into HCs by regulating the mTOR pathway.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , Signal Transduction , TOR Serine-Threonine Kinases , Ubiquitin Thiolesterase , Animals , Cell Transdifferentiation/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Indoles , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/cytology , Oximes , TOR Serine-Threonine Kinases/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Rats
6.
Stem Cells Transl Med ; 13(7): 661-677, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38709826

ABSTRACT

Loss of cochlear hair cells (HCs) leads to permanent hearing loss in mammals, and regenerative medicine is regarded as an ideal strategy for hearing recovery. Limited genetic and pharmaceutical approaches for HC regeneration have been established, and the existing strategies cannot achieve recovery of auditory function. A promising target to promote HC regeneration is MEK/ERK signaling because dynamic shifts in its activity during the critical stages of inner ear development have been observed. Here, we first showed that MEK/ERK signaling is activated specifically in supporting cells (SCs) after aminoglycoside-induced HC injury. We then selected 4 MEK/ERK signaling inhibitors, and PD0325901 (PD03) was found to induce the transdifferentiation of functional supernumerary HCs from SCs in the neonatal mammalian cochlear epithelium. We next found that PD03 facilitated the generation of HCs in inner ear organoids. Through genome-wide high-throughput RNA sequencing and verification, we found that the Notch pathway is the downstream target of MEK/ERK signaling. Importantly, delivery of PD03 into the inner ear induced mild HC regeneration in vivo. Our study thus reveals the importance of MEK/ERK signaling in cell fate determination and suggests that PD03 might serve as a new approach for HC regeneration.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , MAP Kinase Signaling System , Receptors, Notch , Animals , Cell Transdifferentiation/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , MAP Kinase Signaling System/drug effects , Mice , Receptors, Notch/metabolism , Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Labyrinth Supporting Cells/metabolism
7.
Mol Biol Rep ; 51(1): 675, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38787484

ABSTRACT

BACKGROUND: Bioscaffolds and cells are two main components in the regeneration of damaged tissues via cell therapy. Umbilical cord stem cells are among the most well-known cell types for this purpose. The main objective of the present study was to evaluate the effect of the pretreatment of the foreskin acellular matrix (FAM) by monophosphoryl lipid A (MPLA) and Lactobacillus casei supernatant (LCS) on the attraction of human umbilical cord mesenchymal stem cells (hucMSC). METHODS AND RESULTS: The expression of certain cell migration genes was studied using qRT-PCR. In addition to cell migration, transdifferentiation of these cells to the epidermal-like cells was evaluated via immunohistochemistry (IHC) and immunocytochemistry (ICC) of cytokeratin 19 (CK19). The hucMSC showed more tissue tropism in the presence of MPLA and LCS pretreated FAM compared to the untreated control group. We confirmed this result by scanning electron microscopy (SEM) analysis, glycosaminoglycan (GAG), collagen, and DNA content. Furthermore, IHC and ICC data demonstrated that both treatments increase the protein expression level of CK19. CONCLUSION: Pretreatment of acellular bioscaffolds by MPLA or LCS can increase the migration rate of cells and also transdifferentiation of hucMSC to epidermal-like cells without growth factors. This strategy suggests a new approach in regenerative medicine.


Subject(s)
Lacticaseibacillus casei , Lipid A , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Lacticaseibacillus casei/metabolism , Lipid A/metabolism , Lipid A/analogs & derivatives , Cell Movement/drug effects , Skin/metabolism , Tissue Scaffolds/chemistry , Male , Umbilical Cord/cytology , Umbilical Cord/metabolism , Foreskin/cytology , Cell Transdifferentiation/drug effects , Tissue Engineering/methods , Extracellular Matrix/metabolism , Keratin-19/metabolism , Keratin-19/genetics
8.
Exp Cell Res ; 421(1): 113374, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36206825

ABSTRACT

Renal fibrosis is a global health concern with limited curative treatment. Canonical transient receptor potential channel 6 (TRPC6), a nonselective cation channel, has been shown to regulate the renal fibrosis in murine models. However, the molecular mechanism is unclear. Fibroblast-myofibroblast transdifferentiation is one of the critical steps in the progression of renal fibrosis. In the present study, we demonstrate that transforming growth factor (TGF)-ß1 exposure significantly increases the TRPC6 expression in renal interstitial fibroblast NRK-49F cells. Pharmacological inhibition of TRPC6 and knockdown of Trpc6 by siRNA alleviate TGF-ß1-increased expression levels of α-smooth muscle actin (α-SMA) and collagen I, two key markers of myofibroblasts. Although direct activation of TRPC6 by 1-oleoyl-2-acetyl-sn-glycerol (OAG) does not affect the expression of α-SMA and collagen I, OAG potentiates TGF-ß1-induced fibroblast-myofibroblast transdifferentiation. Further study demonstrates that TGF-ß1 exposure increases the phosphorylation level of p38 and Yes-associated protein (YAP) translocation into the nuclei. Inhibition of p38 and YAP decreases TGF-ß1-enhanced TRPC6 and α-SMA expression. In conclusion, we demonstrate that TRPC6 is a key regulator of TGF-ß1-induced fibroblast-myofibroblast transdifferentiation and provides the mechanism of how TGF-ß1 exposure regulates TRPC6 expression in NRK-49F fibroblasts.


Subject(s)
Cell Transdifferentiation , Kidney Diseases , TRPC6 Cation Channel , Animals , Mice , Actins/metabolism , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/physiology , Collagen Type I/metabolism , Fibroblasts/metabolism , Fibrosis , Kidney Diseases/metabolism , Myofibroblasts/metabolism , RNA, Small Interfering/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factors/metabolism , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/therapeutic use , TRPC6 Cation Channel/antagonists & inhibitors , TRPC6 Cation Channel/genetics , YAP-Signaling Proteins , Rats , Disease Models, Animal
9.
J Neuroimmunol ; 364: 577806, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35121334

ABSTRACT

Parkinson's disease (PD) is a chronic and progressive movement disorder caused by the selective loss of midbrain dopaminergic neurons of unknown etiology. Up to now, although there is a great development on treatments of PD, cures with neuroprotective or nerve regenerative effects are underway for PD patients. Here we reported neuroprotective effects of Ginkgolide K (GK) when mice were upon acute MPTP exposure, in which GK ameliorated the gait dysfunction and dopaminergic neuron loss. GK exhibits its ability in immunomodulation, including switching microglia to M2 phenotype and decreasing the microglia-mediated inflammation, inhibiting peripheral CD4+IFN-γ+ and CD4+IL-17+ T cells and α-synuclein specific autoantibodies. The expression of neurotrophic factors BDNF, GDNF and NT-3 was promoted with a treatment of GK in MPTP mice brains. Notably, GK enhanced the expression of nestin in GFAP+ astrocytes followed by the transdifferentiation of astrocyte-to-neuron independent on the Wnt signaling although GK induced the expression of Wnt signaling on astrocytes. Based on these results, our work implicates a therapeutic potential of GK for protecting TH+ neurons by multiple and intercellular pathways to modify nerve regeneration in MPTP mice. However, its exactly cellular and molecular mechanisms need to be further explored and confirmed.


Subject(s)
Astrocytes/drug effects , Cell Transdifferentiation/drug effects , Dopaminergic Neurons , Ginkgolides/pharmacology , Lactones/pharmacology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders , Animals , Male , Mice , Mice, Inbred C57BL
10.
Diabetes ; 71(2): 249-263, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34732538

ABSTRACT

The increasing prevalence of obesity has resulted in demands for the development of new effective strategies for obesity treatment. Withaferin A (WA) shows a great potential for prevention of obesity by sensitizing leptin signaling in the hypothalamus. However, the mechanism underlying the weight- and adiposity-reducing effects of WA remains to be elucidated. In this study, we report that WA treatment induced white adipose tissue (WAT) browning, elevated energy expenditure, decreased respiratory exchange ratio, and prevented high-fat diet-induced obesity. The sympathetic chemical denervation dampened the WAT browning and also impeded the reduction of adiposity in WA-treated mice. WA markedly upregulated the levels of Prdm16 and FATP1 (Slc27a1) in the inguinal WAT (iWAT), and this was blocked by sympathetic denervation. Prdm16 or FATP1 knockdown in iWAT abrogated the WAT browning-inducing effects of WA and restored the weight gain and adiposity in WA-treated mice. Together, these findings suggest that WA induces WAT browning through the sympathetic nerve-adipose axis, and the adipocytic Prdm16-FATP1 pathway mediates the promotive effects of WA on white adipose browning.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Obesity/prevention & control , Withanolides/pharmacology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/physiology , Adipose Tissue, White/innervation , Adipose Tissue, White/physiology , Animals , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
11.
J Endocrinol Invest ; 45(1): 95-103, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34191257

ABSTRACT

AIMS: Metformin, rosiglitazone and sulfonylureas enhance either insulin action or secretion and thus have been used extensively as early stage anti-diabetic medication, independently of the aetiology of the disease. When administered to newly diagnosed diabetes patients, these drugs produce variable results. Here, we examined the effects of the three early stage oral hypoglycaemic agents in mice with diabetes induced by multiple low doses of streptozotocin, focusing specifically on the developmental biology of pancreatic islets. METHODS: Streptozotocin-treated diabetic mice expressing a fluorescent reporter specifically in pancreatic islet α-cells were administered the biguanide metformin (100 mg/kg), thiazolidinedione rosiglitazone (10 mg/kg), or sulfonylurea tolbutamide (20 mg/kg) for 10 days. We assessed the impact of the treatment on metabolic status of the animals as well as on the morphology, proliferative potential and transdifferentiation of pancreatic islet cells, using immunofluorescence. RESULTS: The effect of the therapy on the islet cells varied depending on the drug and included enhanced pancreatic islet ß-cell proliferation, in case of metformin and rosiglitazone; de-differentiation of α-cells and ß-cell apoptosis with tolbutamide; increased relative number of ß-cells and bi-hormonal insulin + glucagon + cells with metformin. These effects were accompanied by normalisation of food and fluid intake with only minor effects on glycaemia at the low doses of the agents employed. CONCLUSIONS: Our data suggest that metformin and rosiglitazone attenuate the depletion of the ß-cell pool in the streptozotocin-induced diabetes, whereas tolbutamide exacerbates the ß-cell apoptosis, but is likely to protect ß-cells from chronic hyperglycaemia by directly elevating insulin secretion.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Insulin Secretion/drug effects , Islets of Langerhans , Metformin/pharmacology , Rosiglitazone/pharmacology , Animals , Blood Glucose/metabolism , Cell Differentiation/drug effects , Cell Transdifferentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice
12.
PLoS One ; 16(12): e0261608, 2021.
Article in English | MEDLINE | ID: mdl-34929019

ABSTRACT

Ac3IV (Ac-CYIQNCPRG-NH2) is an enzymatically stable vasopressin analogue that selectively activates Avpr1a (V1a) and Avpr1b (V1b) receptors. In the current study we have employed streptozotocin (STZ) diabetic transgenic Ins1Cre/+;Rosa26-eYFP and GluCreERT2;Rosa26-eYFP mice, to evaluate the impact of sustained Ac3IV treatment on pancreatic islet cell morphology and transdifferentiation. Twice-daily administration of Ac3IV (25 nmol/kg bw) to STZ-diabetic Ins1Cre/+;Rosa26-eYFP mice for 12 days increased pancreatic insulin (p<0.01) and significantly reversed the detrimental effects of STZ on pancreatic islet morphology. Such benefits were coupled with increased (p<0.01) beta-cell proliferation and decreased (p<0.05) beta-cell apoptosis. In terms of islet cell lineage tracing, induction of diabetes increased (p<0.001) beta- to alpha-cell differentiation in Ins1Cre/+;Rosa26-eYFP mice, with Ac3IV partially reversing (p<0.05) such transition events. Comparable benefits of Ac3IV on pancreatic islet architecture were observed in STZ-diabetic GluCreERT2;ROSA26-eYFP transgenic mice. In this model, Ac3IV provoked improvements in islet morphology which were linked to increased (p<0.05-p<0.01) transition of alpha- to beta-cells. Ac3IV also increased (p<0.05-p<0.01) CK-19 co-expression with insulin in pancreatic ductal and islet cells. Blood glucose levels were unchanged by Ac3IV in both models, reflecting the severity of diabetes induced. Taken together these data indicate that activation of islet receptors for V1a and V1b positively modulates alpha- and beta-cell turnover and endocrine cell lineage transition events to preserve beta-cell identity and islet architecture.


Subject(s)
Cell Transdifferentiation/drug effects , Glucagon-Secreting Cells/drug effects , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Vasopressins/pharmacology , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/drug therapy , Glucagon/metabolism , Glucagon-Secreting Cells/pathology , Insulin/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/anatomy & histology , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Vasopressin
13.
PLoS One ; 16(11): e0256812, 2021.
Article in English | MEDLINE | ID: mdl-34762649

ABSTRACT

Transforming growth factor-beta 1 (TGF-ß1), a pro-fibrotic tumour-derived factor promotes fibroblast differentiation in the tumour microenvironment and is thought to contribute to the development of pro-tumourigenic cancer-associated fibroblasts (CAFs) by promoting myofibroblast differentiation. miRNA dysregulation has been demonstrated in myofibroblast transdifferentiation and CAF activation, however, their expression varies among cell types and with the method of fibroblast induction. Here, the expression profile of miRNA in human primary oral fibroblasts treated with TGF-ß1, to derive a myofibroblastic, CAF-like phenotype, was determined compared to untreated fibroblasts. Myofibroblast transdifferentiation was determined by the expression of alpha-smooth muscle actin (α-SMA) and fibronectin-1 extra domain A (FN-EDA1) using quantitative real-time PCR (qRT-PCR) and western blot. The formation of stress fibres was assessed by fluorescence microscopy, and associated changes in contractility were assessed using collagen contraction assays. Extracellular vesicles (EVs) were purified by using size exclusion chromatography and ultracentrifugation and their size and concentration were determined by nanoparticle tracking analysis. miRNA expression profiling in oral fibroblasts treated with TGF-ß1 and their extracellular vesicles was carried out using tiling low-density array cards. The Database for Annotation, Visualization, and Integrated Discovery (DAVID) was used to perform functional and pathway enrichment analysis of target genes. In this study, TGF-ß1 induced a myofibroblastic phenotype in normal oral fibroblasts as assessed by expression of molecular markers, the formation of stress fibres and increased contractility. TaqMan Low-Density Array (TLDA) analysis demonstrated that miR-503 and miR-708 were significantly upregulated, while miR-1276 was significantly downregulated in TGF-ß1-treated oral fibroblasts (henceforth termed experimentally-derived CAF, eCAF). The gene functional enrichment analysis showed that the candidate miRNAs have the potential to modulate various pathways; including the Ras associated protein 1 (Rap1), PI3K-Akt, and tumour necrosis factor (TNF) signalling pathways. In addition, altered levels of several miRNAs were detected in eCAF EV, including miR-142 and miR-222. No differences in size or abundance of EV were detected between eCAF and normal oral fibroblast (NOF). Little overlap was observed between changes in cellular and EV miRNA profiles, suggesting the possibility of selective loading of EV miRNA. The study reveals miRNA expression signature could be involved in myofibroblast transdifferentiation and the miRNA cargo of their EV, providing novel insight into the involvement of miRNA in CAF development and function.


Subject(s)
Cell Transdifferentiation/physiology , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Myofibroblasts/cytology , Actins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Collagen/metabolism , Extracellular Vesicles/drug effects , Gene Expression Profiling , Humans , MicroRNAs/genetics , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta1/pharmacology
14.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769044

ABSTRACT

Arterial media calcification (AMC) is predominantly regulated by vascular smooth muscle cells (VSMCs), which transdifferentiate into pro-calcifying cells. In contrast, there is little evidence for endothelial cells playing a role in the disease. The current study investigates cellular functioning and molecular pathways underlying AMC, respectively by, an ex vivo isometric organ bath set-up to explore the interaction between VSMCs and ECs and quantitative proteomics followed by functional pathway interpretation. AMC development, which was induced in mice by dietary warfarin administration, was proved by positive Von Kossa staining and a significantly increased calcium content in the aorta compared to that of control mice. The ex vivo organ bath set-up showed calcified aortic segments to be significantly more sensitive to phenylephrine induced contraction, compared to control segments. This, together with the fact that calcified segments as compared to control segments, showed a significantly smaller contraction in the absence of extracellular calcium, argues for a reduced basal NO production in the calcified segments. Moreover, proteomic data revealed a reduced eNOS activation to be part of the vascular calcification process. In summary, this study identifies a poor endothelial function, next to classic pro-calcifying stimuli, as a possible initiator of arterial calcification.


Subject(s)
Endothelial Cells/pathology , Tunica Media/drug effects , Vascular Calcification/chemically induced , Vascular Calcification/pathology , Warfarin/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Calcification, Physiologic/drug effects , Calcium/metabolism , Cell Transdifferentiation/drug effects , Endothelial Cells/metabolism , Male , Mice , Mice, Inbred DBA , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Osteogenesis/drug effects , Tunica Media/metabolism , Tunica Media/pathology , Vascular Calcification/metabolism
15.
Mediators Inflamm ; 2021: 3399357, 2021.
Article in English | MEDLINE | ID: mdl-34690551

ABSTRACT

Liver fibrosis, which results from chronic liver injury due to factors such as chronic alcohol consumption, hepatitis virus infections, and immune attacks, is marked by excessive deposition of extracellular matrix (ECM). Resveratrol (Res), a polyphenol phytoalexin, has been demonstrated to show anti-inflammatory, antioxidative, antiproliferative, and chemopreventive activities. In recent years, Res has been found to inhibit liver fibrosis. Enhanced Hippo pathway activation has also been reported to inhibit tumor progression and liver fibrosis. In the present study, the role of the Hippo pathway in mediating the effects of Res on hepatic stellate cells (HSCs) was examined. We found that Res significantly suppresses HSC proliferation, reducing the cell index. Res induced HSC inactivation, reducing collagen deposition and α-smooth muscle actin (α-SMA) expression. In addition, Res contributed to HSC apoptosis, upregulating Bax and downregulating Bcl-2 expression. Notably, the Hippo pathway was involved in the Res-mediated suppression of HSC activation. Res enhanced the activation of the Hippo pathway and reduced yes-associated protein (YAP) and transcriptional coactivator with the PDZ-binding motif (TAZ) expression. Interestingly, the YAP overexpression inhibited Res-induced HSC inactivation and apoptosis. In conclusion, these results demonstrate that Res inhibits HSC activation, at least in part, via the Hippo pathway. The present study indicates a new antifibrotic mechanism of Res and provides novel insights into Hippo-mediated HSC apoptosis and HSC activation in liver fibrosis.


Subject(s)
Hepatic Stellate Cells/drug effects , Hippo Signaling Pathway/drug effects , Resveratrol/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Cells, Cultured , Collagen/metabolism , Hepatic Stellate Cells/physiology , Hippo Signaling Pathway/physiology , Humans , Liver Cirrhosis/drug therapy , Male , Mice , Mice, Inbred C57BL , Resveratrol/therapeutic use , YAP-Signaling Proteins/physiology
16.
Cell Rep ; 37(1): 109774, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610301

ABSTRACT

While squamous transdifferentiation within subpopulations of adenocarcinomas represents an important drug resistance problem, its underlying mechanism remains poorly understood. Here, using surface markers of resistant basal cell carcinomas (BCCs) and patient single-cell and bulk transcriptomic data, we uncover the dynamic roadmap of basal to squamous cell carcinoma transition (BST). Experimentally induced BST identifies activator protein 1 (AP-1) family members in regulating tumor plasticity, and we show that c-FOS plays a central role in BST by regulating the accessibility of distinct AP-1 regulatory elements. Remarkably, despite prominent changes in cell morphology and BST marker expression, we show using inducible model systems that c-FOS-mediated BST demonstrates reversibility. Blocking EGFR pathway activation after c-FOS induction partially reverts BST in vitro and prevents BST features in both mouse models and human tumors. Thus, by identifying the molecular basis of BST, our work reveals a therapeutic opportunity targeting plasticity as a mechanism of tumor resistance.


Subject(s)
Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Cell Transdifferentiation , Proto-Oncogene Proteins c-fos/metabolism , Animals , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/veterinary , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/veterinary , Cell Transdifferentiation/drug effects , Chromatin Assembly and Disassembly , Drug Resistance, Neoplasm/genetics , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mucin-1/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/genetics , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , ras Proteins/genetics , ras Proteins/metabolism
17.
Biomed Pharmacother ; 144: 112284, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34626932

ABSTRACT

Hepatic fibrosis is a wound-healing process caused by prolonged liver damage and often occurs due to hepatic stellate cell activation in response to reactive oxygen species (ROS). Red raspberry has been found to attenuate oxidative stress, mainly because it is rich in bioactive components. In the current study, we investigated the inhibitory effects and associated molecular mechanisms of red raspberry extract (RBE) upon activated hepatic stellate cell (aHSC) in cellular and rat models. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were increased in the dimethylnitrosamine (DMN)-applied samples, whereas treatment of RBE significantly suppressed the activities of these enzymes. In addition, a histopathological analysis demonstrated that RBE could substantially diminish the hepatic collagen content and alpha-smooth muscle actin (α-SMA) expression induced by DMN. Administration of 250 µg/mL RBE could also arrest the growth and enhance the apoptosis of activated HSC-T6 cells, which was accompanied with elevated levels of activated caspases and poly (ADP-ribose) polymerase (PARP) cleavage. Particularly, RBE application remarkably abolished oxidative damage within the cells and reduced the carbonylation of proteins, which was attributed to the upregulation of catalase, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1). Moreover, the knockdown of Nrf2 together with the RBE treatment synergistically abrogated the expression of α-SMA and promoted the level of peroxisome proliferator-activated receptor gamma (PPAR-γ), suggesting that RBE could mitigate the transdifferentiation of HSC in a Nrf2-independent manner. These findings implied that the application of RBE could effectively remove oxidative stress and relieve the activation of HSC via modulating the caspase/PARP, Nrf2/HO-1 and PPAR-γ pathways, which may allow the development of novel therapeutic strategies against chemical-caused liver fibrogenesis.


Subject(s)
Antifibrotic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Transdifferentiation/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/prevention & control , Liver/drug effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Rubus , Animals , Antifibrotic Agents/isolation & purification , Antioxidants/isolation & purification , Apoptosis Regulatory Proteins/metabolism , Cell Line , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Fruit , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , PPAR gamma/metabolism , Plant Extracts/isolation & purification , Protein Carbonylation/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism , Rubus/chemistry , Signal Transduction
18.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34606582

ABSTRACT

Endometriosis is characterized by inflammation and fibrotic changes. Our previous study using a mouse model showed that proinflammatory factors present in peritoneal hemorrhage exacerbated inflammation in endometriosis-like grafts, at least in part through the activation of prostaglandin (PG) E2 receptor and protease-activated receptor (PAR). In addition, menstruation-related factors, PGE2 and thrombin (P/T), a PAR1 agonist induced epithelial-mesenchymal transition (EMT) of endometrial cells under hypoxia. However, the molecular mechanisms by which P/T induce development of endometriosis have not been fully characterized. To investigate the effects of P/T, RNA extracted from endometrial stromal cells (ESCs) treated with P/T were subjected to RNA sequence analysis, and identified activin A, FOS, and GATA2 as upregulated genes. Activin A increased the expression of connective tissue growth factor (CTGF) and mesenchymal marker genes in ESCs. CTGF induced the expression of fibrosis marker type I collagen, fibronectin, and α-smooth muscle actin (αSMA), indicating fibroblast to myofibroblast transdifferentiation (FMT) of ESCs. In addition, activin A, FOS, GATA2, CTGF, and αSMA were localized in endometriosis lesions. Taken together, our data show that P/T induces changes resembling EMT and FMT in ectopic ESCs derived from retrograde menstruation, and that these are associated with fibrotic changes in the lesions. Pharmacological means that block P/T-induced activin A and CTGF signaling may be strategies to inhibit fibrosis in endometriotic lesions.


Subject(s)
Cell Transdifferentiation/drug effects , Dinoprostone/pharmacology , Endometrium/drug effects , Myofibroblasts/drug effects , Thrombin/pharmacology , Activins/genetics , Activins/metabolism , Adult , Cell Transdifferentiation/genetics , Cells, Cultured , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Endometriosis/pathology , Endometrium/cytology , Endometrium/pathology , Female , Humans , Myofibroblasts/physiology , Peritoneal Diseases/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Stromal Cells/drug effects , Stromal Cells/pathology , Stromal Cells/physiology
19.
PLoS One ; 16(9): e0249438, 2021.
Article in English | MEDLINE | ID: mdl-34473703

ABSTRACT

Muscle derived stem cells (MDSCs) and myoblast play an important role in myotube regeneration when muscle tissue is injured. However, these cells can be induced to differentiate into adipocytes once exposed to PPARγ activator like EPA and DHA that are highly suggested during pregnancy. The objective of this study aims at determining the identity of trans-differentiated cells by exploring the effect of EPA and DHA on C2C12 undergoing differentiation into brown and white adipocytes. DHA but not EPA committed C2C12 cells reprograming into white like adipocyte phenotype. Also, DHA promoted the expression of lipolysis regulating genes but had no effect on genes regulating ß-oxidation referring to its implication in lipid re-esterification. Furthermore, DHA impaired C2C12 cells differentiation into brown adipocytes through reducing the thermogenic capacity and mitochondrial biogenesis of derived cells independent of UCP1. Accordingly, DHA treated groups showed an increased accumulation of lipid droplets and suppressed mitochondrial maximal respiration and spare respiratory capacity. EPA, on the other hand, reduced myogenesis regulating genes, but no significant differences were observed in the expression of adipogenesis key genes. Likewise, EPA suppressed the expression of WAT signature genes indicating that EPA and DHA have an independent role on white adipogensis. Unlike DHA treatment, EPA supplementation had no effect on the differential of C2C12 cells into brown adipocytes. In conclusion, DHA is a potent adipogenic and lipogenic factor that can change the metabolic profile of muscle cells by increasing myocellular fat.


Subject(s)
Adipocytes, White/drug effects , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Adipocytes, Brown/drug effects , Adipocytes, White/cytology , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Animals , Cell Line , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , DNA, Mitochondrial , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipolysis/drug effects , Mice , Myoblasts/cytology , Myoblasts/drug effects
20.
Basic Clin Pharmacol Toxicol ; 129(6): 462-469, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34571584

ABSTRACT

Keloid is a type of unusually raised scar. Botulinum toxin A (BTX-A) has a great application potential in keloids treatment. Here, we investigated the functional role of BTX-A in keloids. We separated keloid tissues and normal skin tissues from keloid patients and found that the expression of myofibroblast markers, α-SMA, Collagen I, and Collagen III was increased in the keloid tissues as compared with normal skin tissues. Keloid fibroblasts derived from keloid tissues were treated with TGF-ß1 to induce the differentiation of fibroblasts into myofibroblasts. The keloid myofibroblasts displayed a significant up-regulation of α-SMA. BTX-A enhanced the expression of adipocyte markers, PPARγ and C/EBPα, and increased the accumulation of lipid droplets, and reduced the expression of α-SMA, Collagen I, and Collagen III in the keloid myofibroblasts. Moreover, BTX-A enhanced the expression of BMP4 and p-smad1/5/8. Noggin (BMP4 antagonist) treatment reversed BTX-A-mediated increase of PPARγ and C/EBPα expression and lipid droplets, and down-regulation of α-SMA, Collagen I, and Collagen III in primary keloid myofibroblasts. In conclusion, BTX-A promoted the transdifferentiation of primary keloid myofibroblasts into adipocyte-like cells, which may attribute to activate BMP4/Smad signalling pathway. Thus, this study provides new insights into the mechanism of BTX-A in keloid.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Keloid/drug therapy , Myofibroblasts/drug effects , Neuromuscular Agents/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Bone Morphogenetic Protein 4/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Collagen Type III/metabolism , Fibroblasts/cytology , Humans , Keloid/pathology , Myofibroblasts/cytology , Signal Transduction/drug effects , Smad Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL