Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 441
Filter
1.
Tissue Eng Part A ; 30(7-8): 330-339, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37819701

ABSTRACT

Vascular endothelial cells (ECs), locating at the inner side of vascular lumen, play critical roles in maintaining vascular function and participate in tissue repair and neovascularization. Although increasing studies have shown positive effects of transplantation of vascular ECs or their precursor cells on neovascularization and functional recovery of ischemic tissues, the quantity of in vivo ECs is limited and their quality is affected by age, gender, disease, and others, which hinder their clinical application and further study. Chemical transdifferentiation is a promising approach to generate patient-specific cells. In this process, somatic cells are directly converted into desired cell types without the risk of tumorigenicity by pluripotent cell transplantation and exogenous gene introduction by transgene technology. In the present study, we derived ECs from human cardiac fibroblasts (CFs) through an optimized chemical induction method. The derived ECs expressed endothelial specific markers, took up low-density lipoprotein, secreted angiogenic cytokines under hypoxic condition, and formed microvessels in vitro and in vivo. This CF-EC transition bypassed pluripotency and germ layer differentiation, but underwent a stage of endothelialization. Although p53 maintained the same level during the period of CF-EC transdifferentiation, we could modulate p53 transcriptional activity to further improve cell transition efficiency, which mainly functioned at the later stage of endothelialization. Optimization and exploring the regulatory mechanism of CF-EC transition complement each other, which not only broadens the sources of patient-specific ECs but also provides valuable references for the in vivo direct transdifferentiation study and the elucidation of endothelial development and dysfunction. Impact statement This study provides an optimized chemical induction method to derive endothelial cells (ECs) from human cardiac fibroblasts (CFs), which not only broadens the sources of patient-specific ECs but also provides a good research model of mesenchymal-endothelial transition. Studying the molecular process and regulatory mechanism of CF-EC transdifferentiation will provide valuable references for the in vivo direct transdifferentiation for clinical therapy and deepen the understanding of endothelial development and dysfunction.


Subject(s)
Cell Transdifferentiation , Endothelial Cells , Humans , Cell Transdifferentiation/physiology , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , Fibroblasts , Cell Differentiation/physiology , Neovascularization, Physiologic
2.
Curr Opin Cell Biol ; 85: 102255, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37806296

ABSTRACT

The hematopoietic system is one of the earliest tissues to develop. De novo generation of hematopoietic progenitor and stem cells occurs through a transdifferentiation of (hemogenic) endothelial cells to hematopoietic identity, resulting in the formation of intra-aortic hematopoietic cluster (IAHC) cells. Heterogeneity of IAHC cell phenotypes and functions has stymied the field in its search for the transcriptional program of emerging hematopoietic stem cells (HSCs), given that an individual IAHC cannot be simultaneously examined for function and transcriptome. Several models could account for this heterogeneity, including a novel model suggesting that the transcriptomes of individual emerging IAHC cells are in an unstable/metastable state, with pivotal hematopoietic transcription factors expressed dynamically due to transcriptional pulsing and combinatorial activities. The question remains - how is functional hematopoietic cell fate established - is the process stochastic? This article touches upon these important issues, which may be relevant to the field's inability to make HSCs ex vivo.


Subject(s)
Endothelial Cells , Hematopoietic Stem Cells , Cell Differentiation , Cell Transdifferentiation/physiology , Stochastic Processes
3.
Cell Signal ; 110: 110812, 2023 10.
Article in English | MEDLINE | ID: mdl-37468053

ABSTRACT

Portal hypertension is a group of clinical syndromes induced by increased portal system pressure due to various etiologies including cirrhosis. When portal hypertension develops, the portal vein dilates and endothelial cells (ECs) in the portal vein are subjected to mechanical stretch. In this study, elastic silicone chambers were used to simulate the effects of mechanical stretch on ECs under portal hypertension. We found that mechanical stretch decreased PPARγ expression in ECs by blocking the PI3K/AKT/CREB signaling pathway or increasing NEDD4-mediated ubiquitination and degradation of PPARγ. Moreover, PPARγ downregulation triggered Endothelial-to-mesenchymal transition (EndoMT) in ECs under stretch by promoting Smad3 phosphorylation. The PPARγ agonist rosiglitazone mitigated stretch-induced EndoMT in vitro and alleviated EndoMT of the portal vein endothelium in cirrhotic rats.


Subject(s)
Cell Transdifferentiation , Endothelial Cells , Hypertension, Portal , Animals , Rats , Down-Regulation , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hypertension, Portal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , PPAR gamma/metabolism , Stress, Mechanical , Cell Transdifferentiation/physiology
4.
Cell Prolif ; 56(5): e13422, 2023 May.
Article in English | MEDLINE | ID: mdl-36786003

ABSTRACT

Both exogenous transcriptional factors and chemical-defined medium can transdifferentiate astrocytes into functional neurons. However, the regional preference for such transdifferentiation has not been fully studied. A previously reported 5C medium was infused into the mouse cortex and striatum to determine the regional preference for transdifferentiation from astrocytes to neurons. The numbers of NeuN+ GFAP+ EdU+ cells (intermediates) and NeuN+ EdU+ cells (end products) were determined by immunofluorescence to explore the regional preference of transdifferentiation. In addition, to optimize the delivery of the transdifferentiation medium, three key growth factors, insulin, bFGF and transferrin, were loaded onto chitosan nanoparticles, mixed with gelatin methacryloyl and tested in animals with motor cortex injury. A higher transdifferentiation efficiency was identified in the mouse cortex. Differences in cellular respiration and the balance between glutaminase (Gls) and glutamine synthetase were confirmed to be key regulators. In addition, the sustained drug release system induced transdifferentiation of cortex astrocytes both in vivo and in vitro, and partially facilitated the behaviour recovery of mice with motor cortex injury. We also applied this method in pigs and obtained consistent results. In summary, low Gls and glycolysis can be used to predict high transdifferentiation efficiency, which may be useful to identify better indications for the current transdifferentiation system. In addition, the current drug delivery system has the potential to treat diseases related to cortex injuries.


Subject(s)
Cell Transdifferentiation , Glutaminase , Mice , Animals , Swine , Cell Transdifferentiation/physiology , Glutaminase/metabolism , Cells, Cultured , Astrocytes/metabolism , Glycolysis
5.
Hepatology ; 77(4): 1198-1210, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36626626

ABSTRACT

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.


Subject(s)
Cholestasis , Liver Diseases , Animals , Zebrafish , Cell Transdifferentiation/physiology , Hepatocytes/metabolism , Liver , Epithelial Cells , Cholestasis/metabolism , Liver Diseases/metabolism , Cell Proliferation , Liver Regeneration/physiology
6.
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
7.
Aging Cell ; 21(7): e13632, 2022 07.
Article in English | MEDLINE | ID: mdl-35653631

ABSTRACT

Cellular senescence is characterized by a stable proliferation arrest in response to stresses and the acquisition of a senescence-associated secretory phenotype, called SASP, composed of numerous factors including pro-inflammatory molecules, proteases, and growth factors. The SASP affects the environment of senescent cells, especially during aging, by inducing and modulating various phenotypes such as paracrine senescence, immune cell activity, and extracellular matrix deposition and organization, which critically impact various pathophysiological situations, including fibrosis and cancer. Here, we uncover a novel paracrine effect of the SASP: the neuroendocrine transdifferentiation (NED) of some epithelial cancer cells, evidenced both in the breast and prostate. Mechanistically, this effect is mediated by NF-κB-dependent SASP factors, and leads to an increase in intracellular Ca2+ levels. Consistently, buffering Ca2+ by overexpressing the CALB1 buffering protein partly reverts SASP-induced NED, suggesting that the SASP promotes NED through a SASP-induced Ca2+ signaling. Human breast cancer dataset analyses support that NED occurs mainly in p53 WT tumors and in older patients, in line with a role of senescent cells and its secretome, as they are increasing during aging. In conclusion, our work, uncovering SASP-induced NED in some cancer cells, paves the way for future studies aiming at better understanding the functional link between senescent cell accumulation during aging, NED and clinical patient outcome.


Subject(s)
Breast Neoplasms , Cell Transdifferentiation , NF-kappa B , Aged , Breast Neoplasms/metabolism , Cell Transdifferentiation/physiology , Cellular Senescence/genetics , Cellular Senescence/physiology , Humans , Male , NF-kappa B/metabolism , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Secretome
8.
Cell Rep ; 38(4): 110295, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081347

ABSTRACT

Genesis of syncytial muscles is typically considered as a paradigm for an irreversible developmental process. Notably, transdifferentiation of syncytial muscles is naturally occurring during Drosophila development. The ventral longitudinal heart-associated musculature (VLM) arises by a unique mechanism that revokes differentiation states of so-called alary muscles and comprises at least two distinct steps: syncytial muscle cell fragmentation into single myoblasts and successive reprogramming into founder cells that orchestrate de novo fiber formation of the VLM lineage. Here, we provide evidence that the mesodermal master regulator twist plays a key role during this reprogramming process. Acting downstream of Drosophila Tbx1 (Org-1), Twist is regulating the activity of the Hippo pathway effector Yorkie and is required for the initiation of syncytial muscle dedifferentiation and fragmentation. Subsequently, fibroblast growth factor receptor (FGFR)-Ras-mitogen-activated protein kinase (MAPK) signaling in resulting mononucleated myoblasts maintains Twist expression, thereby stabilizing nuclear Yorkie activity and inducing their lineage switch into founder cells of the VLM.


Subject(s)
Cellular Reprogramming/physiology , Drosophila Proteins/metabolism , Heart/embryology , Myocardium/cytology , Twist-Related Protein 1/metabolism , YAP-Signaling Proteins/metabolism , Animals , Cell Lineage/physiology , Cell Transdifferentiation/physiology , Drosophila melanogaster
10.
Mol Neurobiol ; 59(2): 983-1001, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34816381

ABSTRACT

With an increase in the incidence of neurodegenerative diseases, a need to replace incapable conventional methods has arisen. To overcome this burden, stem cells therapy has emerged as an efficient treatment option. Endeavours to accomplish this have paved the path to neural regeneration through efficient neuronal transdifferentiation. Despite their potential, the use of stem cells still entails several limitations, such as low differentiation efficiency and difficulties in guiding differentiation. The process of neural differentiation through the stem cells is achieved through the use of chemical inducers or growth factors and their direct introduction reduces their bioavailability in the system. To address these limitations, neural regeneration ventures require growth factors to be effectively implemented on stem cells in order to produce functional neuronal precursor cells. An efficient technique to achieve it is through the delivery of growth factors via microcarriers for their sustained release. It ensures the presence of commensurable concentration even at later stages of neuronal transdifferentiation. Nanofibers and nanoparticles, along with liposomes and such, have been used to implement this. The interaction between such carriers and the growth factors is mainly electrostatic. Such interaction enables them to form a stable assembly through immobilisation of the growth factor either onto their surfaces or within the core of their structures. The rate of sustained release depends upon the release kinetics associated with the polymeric structure employed and its interaction with the encapsulated growth factor. The sustained release ensures that the stem cells immerse under the effect of the growth factors for a prolonged period, ultimately aiding in the formation of cells showing ample characteristics of neuron precursors. This review analyses the various carriers that have been employed for the release of growth factors in an orderly fashion and their constituents, along with the advantages and the limitations they pose in delivering the growth factors for facilitating the process of neuronal transdifferentiation.


Subject(s)
Cell Transdifferentiation , Intercellular Signaling Peptides and Proteins , Mesenchymal Stem Cells , Cell Differentiation , Cell Transdifferentiation/physiology , Delayed-Action Preparations , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Mesenchymal Stem Cells/physiology , Nerve Regeneration
11.
Nat Cell Biol ; 24(1): 10-23, 2022 01.
Article in English | MEDLINE | ID: mdl-34969962

ABSTRACT

Loss of alveolar type 2 cells (AEC2s) and the ectopic appearance of basal cells in the alveoli characterize severe lung injuries such as idiopathic pulmonary fibrosis (IPF). Here we demonstrate that human alveolar type 2 cells (hAEC2s), unlike murine AEC2s, transdifferentiate into basal cells in response to fibrotic signalling in the lung mesenchyme, in vitro and in vivo. Single-cell analysis of normal hAEC2s and mesenchymal cells in organoid co-cultures revealed the emergence of pathologic fibroblasts and basaloid cells previously described in IPF. Transforming growth factor-ß1 and anti-bone morphogenic protein signalling in the organoids promoted transdifferentiation. Trajectory and histologic analyses of both hAEC2-derived organoids and IPF epithelium indicated that hAEC2s transdifferentiate into basal cells through alveolar-basal intermediates that accumulate in proximity to pathologic CTHRC1hi/TGFB1hi fibroblasts. Our study indicates that hAEC2 loss and expansion of alveolar metaplastic basal cells in severe human lung injuries are causally connected through an hAEC2-basal cell lineage trajectory driven by aberrant mesenchyme.


Subject(s)
Cell Transdifferentiation/physiology , Epithelial Cells/cytology , Idiopathic Pulmonary Fibrosis/pathology , Keratin-5/metabolism , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Alveolar Epithelial Cells/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cells, Cultured , Epidermal Cells/cytology , Fibroblasts/cytology , Humans , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Signal Transduction/physiology , Single-Cell Analysis , Transforming Growth Factor beta1/metabolism
12.
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
13.
Dev Cell ; 56(22): 3115-3127.e6, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34562378

ABSTRACT

Acute ischemic stroke damages the regional brain blood vessel (BV) network. Acute recovery of basic blood flows, which is carried out by the earliest regenerated BVs, are critical to improve clinical outcomes and minimize lethality. Although the late-regenerated BVs form via growing along the meninge-derived ingrown lymphatic vessels (iLVs), mechanisms underlying the early, acute BV regeneration remain elusive. Using zebrafish cerebrovascular injury models, we show that the earliest regenerated BVs come from lymphatic transdifferentiation, a hitherto unappreciated process in vertebrates. Mechanistically, the LV-to-BV transdifferentiation occurs exclusively in the stand-alone iLVs through Notch activation. In the track iLVs adhered by late-regenerated BVs, transdifferentiation never occurs because the BV-expressing EphrinB2a paracellularly activates the iLV-expressing EphB4a to inhibit Notch activation. Suppression of LV-to-BV transdifferentiation blocks acute BV regeneration and becomes lethal. These results demonstrate that acute BV regeneration occurs via lymphatic transdifferentiation, suggesting this process and key regulatory molecules EphrinB2a/EphB4a/Notch as new postischemic therapeutic targets.


Subject(s)
Brain Ischemia/physiopathology , Brain/blood supply , Cell Transdifferentiation/physiology , Regeneration/physiology , Animals , Lymphatic System/physiopathology , Lymphatic Vessels/physiology , Meninges/physiopathology , Stroke/physiopathology , Zebrafish
14.
Pflugers Arch ; 473(12): 1899-1910, 2021 12.
Article in English | MEDLINE | ID: mdl-34564739

ABSTRACT

In chronic kidney disease (CKD), hyperphosphatemia promotes medial vascular calcification, a process augmented by osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). VSMC function is regulated by sympathetic innervation, and these cells express α- and ß-adrenergic receptors. The present study explored the effects of ß2-adrenergic stimulation by isoproterenol on VSMC calcification. Experiments were performed in primary human aortic VSMCs treated with isoproterenol during control or high phosphate conditions. As a result, isoproterenol dose dependently up-regulated the expression of osteogenic markers core-binding factor α-1 (CBFA1) and tissue-nonspecific alkaline phosphatase (ALPL) in VSMCs. Furthermore, prolonged isoproterenol exposure augmented phosphate-induced calcification of VSMCs. Isoproterenol increased the activation of PKA and CREB, while knockdown of the PKA catalytic subunit α (PRKACA) or of CREB1 genes was able to suppress the pro-calcific effects of isoproterenol in VSMCs. ß2-adrenergic receptor silencing or inhibition with the selective antagonist ICI 118,551 blocked isoproterenol-induced osteogenic signalling in VSMCs. The present observations imply a pro-calcific effect of ß2-adrenergic overstimulation in VSMCs, which is mediated, at least partly, by PKA/CREB signalling. These observations may support a link between sympathetic overactivity in CKD and vascular calcification.


Subject(s)
Adrenergic Agents/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction/physiology , Vascular Calcification/metabolism , Aorta/metabolism , Calcium/metabolism , Cell Transdifferentiation/physiology , Cells, Cultured , Humans , Osteogenesis/physiology , Phosphates/metabolism , Renal Insufficiency, Chronic/metabolism
15.
Sci Rep ; 11(1): 17764, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493753

ABSTRACT

Endothelial-mesenchymal transition (EndMT) is a form of endothelial dysfunction wherein endothelial cells acquire a mesenchymal phenotype and lose endothelial functions, which contributes to the pathogenesis of intimal hyperplasia and atherosclerosis. The mitogen activated protein kinase 7 (MAPK7) inhibits EndMT and decreases the expression of the histone methyltransferase Enhancer-of-Zeste homologue 2 (EZH2), thereby maintaining endothelial quiescence. EZH2 is the catalytic subunit of the Polycomb Repressive Complex 2 that methylates lysine 27 on histone 3 (H3K27me3). It is elusive how the crosstalk between MAPK7 and EZH2 is regulated in the endothelium and if the balance between MAPK7 and EZH2 is disturbed in vascular disease. In human coronary artery disease, we assessed the expression levels of MAPK7 and EZH2 and found that with increasing intima/media thickness ratio, MAPK7 expression decreased, whereas EZH2 expression increased. In vitro, MAPK7 activation decreased EZH2 expression, whereas endothelial cells deficient of EZH2 had increased MAPK7 activity. MAPK7 activation results in increased expression of microRNA (miR)-101, a repressor of EZH2. This loss of EZH2 in turn results in the increased expression of the miR-200 family, culminating in decreased expression of the dual-specificity phosphatases 1 and 6 who may repress MAPK7 activity. Transfection of endothelial cells with miR-200 family members decreased the endothelial sensitivity to TGFß1-induced EndMT. In endothelial cells there is reciprocity between MAPK7 signaling and EZH2 expression and disturbances in this reciprocal signaling associate with the induction of EndMT and severity of human coronary artery disease.


Subject(s)
Cell Transdifferentiation/physiology , Coronary Artery Disease/pathology , Endothelium, Vascular/pathology , Enhancer of Zeste Homolog 2 Protein/physiology , Mesoderm/pathology , Mitogen-Activated Protein Kinase 7/physiology , Signal Transduction/physiology , Tunica Intima/pathology , 3' Untranslated Regions/genetics , Coronary Artery Disease/enzymology , Coronary Stenosis/enzymology , Coronary Stenosis/pathology , Dual Specificity Phosphatase 1/biosynthesis , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 6/biosynthesis , Dual Specificity Phosphatase 6/genetics , Endothelium, Vascular/enzymology , Enzyme Activation , Gene Expression Regulation , Genes, Reporter , Histone Code , Human Umbilical Vein Endothelial Cells , Humans , Hyperplasia , Mesoderm/enzymology , MicroRNAs/biosynthesis , MicroRNAs/genetics , Tunica Media/pathology
16.
Front Endocrinol (Lausanne) ; 12: 722250, 2021.
Article in English | MEDLINE | ID: mdl-34421829

ABSTRACT

In all forms of diabetes, ß cell mass or function is reduced and therefore the capacity of the pancreatic cells for regeneration or replenishment is a critical need. Diverse lines of research have shown the capacity of endocrine as well as acinar, ductal and centroacinar cells to generate new ß cells. Several experimental approaches using injury models, pharmacological or genetic interventions, isolation and in vitro expansion of putative progenitors followed by transplantations or a combination thereof have suggested several pathways for ß cell neogenesis or regeneration. The experimental results have also generated controversy related to the limitations and interpretation of the experimental approaches and ultimately their physiological relevance, particularly when considering differences between mouse, the primary animal model, and human. As a result, consensus is lacking regarding the relative importance of islet cell proliferation or progenitor differentiation and transdifferentiation of other pancreatic cell types in generating new ß cells. In this review we summarize and evaluate recent experimental approaches and findings related to islet regeneration and address their relevance and potential clinical application in the fight against diabetes.


Subject(s)
Insulin-Secreting Cells/physiology , Pancreas/physiology , Regeneration/physiology , Adult , Animals , Cell Count , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Transdifferentiation/physiology , Humans , Insulin-Secreting Cells/cytology , Mice , Organ Size , Pancreas/cytology , Stem Cells/physiology
17.
Cells ; 10(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34440909

ABSTRACT

Human erythropoietin (EPO) is an N-linked glycoprotein consisting of 166 aa that is produced in the kidney during the adult life and acts both as a peptide hormone and hematopoietic growth factor (HGF), stimulating bone marrow erythropoiesis. EPO production is activated by hypoxia and is regulated via an oxygen-sensitive feedback loop. EPO acts via its homodimeric erythropoietin receptor (EPO-R) that increases cell survival and drives the terminal erythroid maturation of progenitors BFU-Es and CFU-Es to billions of mature RBCs. This pathway involves the activation of multiple erythroid transcription factors, such as GATA1, FOG1, TAL-1, EKLF and BCL11A, and leads to the overexpression of genes encoding enzymes involved in heme biosynthesis and the production of hemoglobin. The detection of a heterodimeric complex of EPO-R (consisting of one EPO-R chain and the CSF2RB ß-chain, CD131) in several tissues (brain, heart, skeletal muscle) explains the EPO pleotropic action as a protection factor for several cells, including the multipotent MSCs as well as cells modulating the innate and adaptive immunity arms. EPO induces the osteogenic and endothelial transdifferentiation of the multipotent MSCs via the activation of EPO-R signaling pathways, leading to bone remodeling, induction of angiogenesis and secretion of a large number of trophic factors (secretome). These diversely unique properties of EPO, taken together with its clinical use to treat anemias associated with chronic renal failure and other blood disorders, make it a valuable biologic agent in regenerative medicine for the treatment/cure of tissue de-regeneration disorders.


Subject(s)
Bone Remodeling/physiology , Cell Transdifferentiation/physiology , Endothelial Cells/cytology , Erythropoiesis/physiology , Erythropoietin/metabolism , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Bone Remodeling/drug effects , Cell Transdifferentiation/drug effects , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Humans , Receptors, Erythropoietin/metabolism , Regenerative Medicine/methods
18.
J Neurosci Res ; 99(10): 2427-2462, 2021 10.
Article in English | MEDLINE | ID: mdl-34259342

ABSTRACT

Astrocytes play essential roles in development, homeostasis, injury, and repair of the central nervous system (CNS). Their development is tightly regulated by distinct spatial and temporal cues during embryogenesis and into adulthood throughout the CNS. Astrocytes have several important responsibilities such as regulating blood flow and permeability of the blood-CNS barrier, glucose metabolism and storage, synapse formation and function, and axon myelination. In CNS pathologies, astrocytes also play critical parts in both injury and repair mechanisms. Upon injury, they undergo a robust phenotypic shift known as "reactive astrogliosis," which results in both constructive and deleterious outcomes. Astrocyte activation and migration at the site of injury provides an early defense mechanism to minimize the extent of injury by enveloping the lesion area. However, astrogliosis also contributes to the inhibitory microenvironment of CNS injury and potentiate secondary injury mechanisms, such as inflammation, oxidative stress, and glutamate excitotoxicity, which facilitate neurodegeneration in CNS pathologies. Intriguingly, reactive astrocytes are increasingly a focus in current therapeutic strategies as their activation can be modulated toward a neuroprotective and reparative phenotype. This review will discuss recent advancements in knowledge regarding the development and role of astrocytes in the healthy and pathological CNS. We will also review how astrocytes have been genetically modified to optimize their reparative potential after injury, and how they may be transdifferentiated into neurons and oligodendrocytes to promote repair after CNS injury and neurodegeneration.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Homeostasis/physiology , Neurogenesis/physiology , Animals , Cell Transdifferentiation/physiology , Central Nervous System/metabolism , Central Nervous System/pathology , Gliosis/metabolism , Gliosis/pathology , Humans
19.
Mol Neurobiol ; 58(10): 5194-5209, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34269964

ABSTRACT

Human menstrual blood-derived mesenchymal stromal cells (MenSCs) have become not only an important source of stromal cells for cell therapy but also a cellular source for neurologic disorders in vitro modeling. By using culture protocols originally developed in our laboratory, we show that MenSCs can be converted into floating neurospheres (NSs) using the Fast-N-Spheres medium for 24-72 h and can be transdifferentiated into functional dopaminergic-like (DALNs, ~ 26% TH + /DAT + flow cytometry) and cholinergic-like neurons (ChLNs, ~ 46% ChAT + /VAChT flow cytometry) which responded to dopamine- and acetylcholine-triggered neuronal Ca2+ inward stimuli when cultured with the NeuroForsk and the Cholinergic-N-Run medium, respectively in a timely fashion (i.e., 4-7 days). Here, we also report a direct transdifferentiation method to induce MenSCs into functional astrocyte-like cells (ALCs) by incubation of MenSCs in commercial Gibco® Astrocyte medium in 7 days. The MSC-derived ALCs (~ 59% GFAP + /S100ß +) were found to respond to glutamate-induced Ca2+ inward stimuli. Altogether, these results show that MenSCs are a reliable source to obtain functional neurogenic cells to further investigate the neurobiology of neurologic disorders.


Subject(s)
Cell Lineage/physiology , Cell Transdifferentiation/physiology , Cholinergic Neurons/physiology , Dopaminergic Neurons/physiology , Menstruation/physiology , Mesenchymal Stem Cells/physiology , Adolescent , Adult , Cells, Cultured , Female , Humans , Young Adult
20.
Dev Cell ; 56(17): 2471-2485.e5, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34331868

ABSTRACT

Adult mammalian tissues such as heart, brain, retina, and the sensory structures of the inner ear do not effectively regenerate, although a latent capacity for regeneration exists at embryonic and perinatal times. We explored the epigenetic basis for this latent regenerative potential in the mouse inner ear and its rapid loss during maturation. In perinatal supporting cells, whose fate is maintained by Notch-mediated lateral inhibition, the hair cell enhancer network is epigenetically primed (H3K4me1) but silenced (active H3K27 de-acetylation and trimethylation). Blocking Notch signaling during the perinatal period of plasticity rapidly eliminates epigenetic silencing and allows supporting cells to transdifferentiate into hair cells. Importantly, H3K4me1 priming of the hair cell enhancers in supporting cells is removed during the first post-natal week, coinciding with the loss of transdifferentiation potential. We hypothesize that enhancer decommissioning during cochlear maturation contributes to the failure of hair cell regeneration in the mature organ of Corti.


Subject(s)
Cell Differentiation/physiology , Hair Cells, Auditory/metabolism , Receptors, Notch/metabolism , Regeneration/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Transdifferentiation/genetics , Cell Transdifferentiation/physiology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Hair Cells, Auditory/cytology , Mice, Transgenic , Regulatory Sequences, Nucleic Acid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...