4,620 results on '"Cell Transdifferentiation"'
Search Results
202. Inhibition of miR-497 Attenuates Oral Submucous Fibrosis by Inhibiting Myofibroblast Transdifferentiation in Buccal Mucosal Fibroblasts
- Author
-
Jie, Huang, Huang, Zhang, Xusheng, Fan, and Junxin, Guo
- Subjects
Transforming Growth Factor beta1 ,MicroRNAs ,Arecoline ,Cell Transdifferentiation ,Mouth Mucosa ,Humans ,Oral Submucous Fibrosis ,Collagen ,Fibroblasts ,Myofibroblasts ,Areca - Abstract
Oral submucous fibrosis (OSF) is a common chronic condition with poor prognosis, and existing therapies for OSF are limited in effectiveness. This study was designed to explore the role of miR-497 in arecoline (AR)-induced OSF.After miR-497 was silenced or overexpressed in buccal mucosa fibroblasts (BMFs), different concentrations of AR (5-200 μg/ml) were applied to incubate BMFs, and 50 μg/ml of AR was chosen for subsequent experiments. Thereafter, collagen gel contraction assay was used to detect the contractile capacity of BMFs. Transwell assay and wound healing assay were applied to detect migration and invasiveness of the cells. In addition, immunofluorescence staining, qRT-PCR and western blot were conducted to measure the expression of miR-497, collagen I and α-SMA, as well as the phosphorylation of Smad2 and Smad3.After successful inhibition or overexpression of miR-497 in AR-induced BMFs, the results showed that miR- 497 inhibition suppressed the contractility, migration and invasiveness of AR-induced BMFs, whereas overexpression of miR-497 produced the opposite. In addition, miR-497 inhibition down-regulated the expression level of collagen I and α-SMA in AR-exposed BMFs. Furthermore, TGF-β1 expression, Smad2 and Smad3 phosphorylation were also repressed in AR-induced BMFs after miR-497 inhibition. Correspondingly, overexpression of miR-497 reversed the expression of the aforementioned proteins.miR-497 inhibition may attenuate OSF by inhibiting myofibroblast transdifferentiation in BMFs via the TGF-β1/Smads signaling pathway, indicating that miR-497 might represent an underlying target for treating OSF.
- Published
- 2022
203. Dysregulated insulin secretion is associated with pancreatic β‐cell hyperplasia and direct acinar‐β‐cell trans‐differentiation in partially <scp>eNOS</scp> ‐deficient mice
- Author
-
Michela Novelli, Matilde Masini, Cecilia Vecoli, Stefania Moscato, Niccola Funel, Anna Pippa, Letizia Mattii, Chiara Ippolito, Daniela Campani, Danilo Neglia, and Pellegrino Masiello
- Subjects
Blood Glucose ,Mice, Knockout ,Hyperplasia ,Physiology ,Diet, High-Fat ,Mice, Inbred C57BL ,Mice ,Glucose ,Insulin-Secreting Cells ,Physiology (medical) ,Cell Transdifferentiation ,Insulin Secretion ,Animals ,Insulin ,Insulin Resistance - Abstract
eNOS-deficient mice were previously shown to develop hypertension and metabolic alterations associated with insulin resistance either in standard dietary conditions (eNOS-/- homozygotes) or upon high-fat diet (HFD) (eNOS+/- heterozygotes). In the latter heterozygote model, the present study investigated the pancreatic morphological changes underlying the abnormal glycometabolic phenotype. C57BL6 wild type (WT) and eNOS+/- mice were fed with either chow or HFD for 16 weeks. After being longitudinally monitored for their metabolic state after 8 and 16 weeks of diet, mice were euthanized and fragments of pancreas were processed for histological, immuno-histochemical and ultrastructural analyses. HFD-fed WT and eNOS+/- mice developed progressive glucose intolerance and insulin resistance. Differently from WT animals, eNOS+/- mice showed a blunted insulin response to a glucose load, regardless of the diet regimen. Such dysregulation of insulin secretion was associated with pancreatic β-cell hyperplasia, as shown by larger islet fractional area and β-cell mass, and higher number of extra-islet β-cell clusters than in chow-fed WT animals. In addition, only in the pancreas of HFD-fed eNOS+/- mice, there was ultrastructural evidence of a number of hybrid acinar-β-cells, simultaneously containing zymogen and insulin granules, suggesting the occurrence of a direct exocrine-endocrine transdifferentiation process, plausibly triggered by metabolic stress associated to deficient endothelial NO production. As suggested by confocal immunofluorescence analysis of pancreatic histological sections, inhibition of Notch-1 signaling, likely due to a reduced NO availability, is proposed as a novel mechanism that could favor both β-cell hyperplasia and acinar-β-cell transdifferentiation in eNOS-deficient mice with impaired insulin response to a glucose load.
- Published
- 2022
- Full Text
- View/download PDF
204. From IL-17 to IFN-γ in inflammatory skin disorders: Is transdifferentiation a potential treatment target?
- Author
-
Arno Belpaire, Nanja van Geel, Reinhart Speeckaert, and Paradowska-Gorycka, Agnieszka
- Subjects
vitiligo ,PSORIASIS ,FEATURES ,Interleukin-17 ,Immunology ,DISTINCT ,IFN and y ,SARCOIDOSIS ,inflammatory skin disease ,INTERLEUKIN-23 ,Interferon-gamma ,IL-17 ,plasticity ,T CELLS ,Cell Transdifferentiation ,VERSUS-HOST-DISEASE ,Medicine and Health Sciences ,Humans ,Psoriasis ,Th17 Cells ,ATOPIC-DERMATITIS ,Immunology and Allergy ,Th17.1 ,Th17 ,TH17 CELLS - Abstract
The targeted inhibition of effector cytokines such as interleukin 17 (IL-17) in psoriasis and IL-13 in atopic dermatitis offers impressive efficacy with a favorable side effect profile. In contrast, the downregulation of interferon gamma (IFN-γ) in T helper (Th) 1-dominant skin disorders may lead to more adverse events, given the crucial role of IFN-γ in antiviral and antitumoral immunity. Modulating Th17 and Th2 cell differentiation is performed by blocking IL-23 and IL-4, respectively, whereas anti-IL-12 antibodies are only moderately effective in downregulating Th1 lymphocyte differentiation. Therefore, a targeted approach of IFN-γ-driven disorders remains challenging. Recent literature suggests that certain pathogenic Th17 cell subsets with Th1 characteristics, such as CD4+CD161+CCR6+CXCR3+IL-17+IFN-y+ (Th17.1) and CD4+CD161+CCR6+CXCR3+IL-17-IFN-y+ (exTh17), are important contributors in Th1-mediated autoimmunity. Differentiation to a Th17.1 or exTh17 profile results in the upregulation of IFN-y. Remarkably, these pathogenic Th17 cell subsets are resistant to glucocorticoid therapy and the dampening effect of regulatory T cells (Treg). The identification of Th17.1/exTh17 cells in auto-immune disorders may explain the frequent treatment failure of conventional immunosuppressants. In this review, we summarize the current evidence regarding the cellular plasticity of Th17 cells in inflammatory skin disorders. A deeper understanding of this phenomenon may lead to better insights into the pathogenesis of various skin diseases and the discovery of a potential new treatment target.
- Published
- 2022
- Full Text
- View/download PDF
205. Human umbilical cord mesenchymal stem cell-derived exosomal miR-335-5p attenuates the inflammation and tubular epithelial–myofibroblast transdifferentiation of renal tubular epithelial cells by reducing ADAM19 protein levels
- Author
-
Zhenhua, Qiu, Zhihui, Zhong, Yuehan, Zhang, Haoling, Tan, Bo, Deng, and Guohuang, Meng
- Subjects
Inflammation ,Epithelial-Mesenchymal Transition ,Medicine (miscellaneous) ,Epithelial Cells ,Mesenchymal Stem Cells ,Cell Biology ,Fibrosis ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Umbilical Cord ,Transforming Growth Factor beta1 ,ADAM Proteins ,MicroRNAs ,Cell Transdifferentiation ,Humans ,Molecular Medicine ,Myofibroblasts - Abstract
Background Renal tubular epithelial–myofibroblast transdifferentiation (EMT) plays a key role in the regulation of renal fibrosis. Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs) play a crucial role in alleviating renal fibrosis and injury. Additionally, hucMSC-derived exosomes contain numerous microRNAs (miRNAs). However, it is unclear whether mesenchymal stem cells can regulate the transforming growth factor (TGF)-β1-induced EMT of human renal tubular epithelial cells (RTECs) through exosomal miRNAs. Method HK-2, a human RTEC line, was co-treated with TGF-β1 and hucMSC-derived exosomes. Additionally, TGF-β1-treated HK-2 cells were transfected with a miR-335-5p mimic and disintegrin and metalloproteinase domain-containing protein 19 (ADAM19)-overexpression plasmid. miR-335-5p expression and ADAM19 protein and inflammation levels were measured via quantitative reverse transcription polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assays, respectively. Results TGF-β1 treatment changed the shape of HK-2 cells from a cobblestone morphology to a long spindle shape, accompanied by an increase in interleukin (IL)-6, tumor necrosis factor-α, IL-1β, collagen I, collagen III, α-smooth muscle actin, vimentin, and N-cadherin protein levels, whereas E-cadherin protein levels were reduced in these HK-2 cells, suggesting that TGF-β1 treatment induced the inflammation and EMT of HK-2 cells. HucMSC-exosomes improved the inflammation and EMT phenotype of TGF-β1-induced HK-2 cells by transferring miR-335-5p. miR-335-5p was found to bind the ADAM19 3′-untranslated region to reduce ADAM19 protein levels. Additionally, miR-335-5p improved the inflammation and EMT phenotype of HK-2 cells by reducing ADAM19 protein levels with TGF-β1 induction. Conclusions HucMSC-derived exosomal miR-335-5p attenuates the inflammation and EMT of HK-2 cells by reducing ADAM19 protein levels upon TGF-β1 induction. This study provides a potential therapeutic strategy and identifies targets for clinically treating renal fibrosis.
- Published
- 2022
- Full Text
- View/download PDF
206. Genomics of sexual cell fate transdifferentiation in the mouse gonad
- Author
-
Mark W Murphy, Micah D Gearhart, Andrew Wheeler, Vivian J Bardwell, and David Zarkower
- Subjects
Male ,Genomics ,Sex Determination Processes ,Gonadal Dysgenesis ,Chromatin ,Mice ,Cell Transdifferentiation ,Testis ,Genetics ,Animals ,Humans ,Female ,Gonads ,Molecular Biology ,Genetics (clinical) ,Transcription Factors - Abstract
Sex determination in mammals hinges on a cell fate decision in the fetal bipotential gonad between formation of male Sertoli cells or female granulosa cells. While this decision normally is permanent, loss of key cell fate regulators such as the transcription factors Dmrt1 and Foxl2 can cause postnatal transdifferentiation from Sertoli to granulosa-like (Dmrt1) or vice versa (Foxl2). Here, we examine the mechanism of male-to-female transdifferentiation in mice carrying either a null mutation of Dmrt1 or a point mutation, R111G, that alters the DNA-binding motif and causes human XY gonadal dysgenesis and sex reversal. We first define genes misexpressed during transdifferentiation and then show that female transcriptional regulators driving transdifferentiation in the mutant XY gonad (ESR2, LRH1, FOXL2) bind chromatin sites related to those normally bound in the XX ovary. We next define gene expression changes and abnormal chromatin compartments at the onset of transdifferentiation that may help destabilize cell fate and initiate the transdifferentiation process. We model the R111G mutation in mice and show that it causes dominant gonadal dysgenesis, analogous to its human phenotype but less severe. We show that R111G partially feminizes the testicular transcriptome and causes dominant disruption of DMRT1 binding specificity in vivo. These data help illuminate how transdifferentiation occurs when sexual cell fate maintenance is disrupted and identify chromatin sites and transcripts that may play key roles in the transdifferentiation process.
- Published
- 2022
207. NF‐κB‐dependent secretome of senescent cells can trigger neuroendocrine transdifferentiation of breast cancer cells
- Author
-
Clotilde Raynard, Xingjie Ma, Anda Huna, Nolwenn Tessier, Amélie Massemin, Kexin Zhu, Jean‐Michel Flaman, Florentin Moulin, Delphine Goehrig, Jean‐Jacques Medard, David Vindrieux, Isabelle Treilleux, Hector Hernandez‐Vargas, Sylvie Ducreux, Nadine Martin, David Bernard, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Yangzhou University, Cardiovasculaire, métabolisme, diabétologie et nutrition (CarMeN), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Hospices Civils de Lyon (HCL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), and ROSSI, Sabine
- Subjects
Male ,Aging ,transdifférenciation neuroendocrinienne ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,cancer du sein ,sénescence cellulaire ,NF-kappa B ,Breast Neoplasms ,Cell Biology ,vieillissement ,Neuroendocrine Cells ,Cell Transdifferentiation ,phénotype sécrétoire associé à la sénescence ,Humans ,Cellular Senescence ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Aged ,Secretome - Abstract
International audience; La sénescence cellulaire est caractérisée par un arrêt stable de la prolifération en réponse aux stress et l'acquisition d'un phénotype sécrétoire associé à la sénescence, appelé SASP, composé de nombreux facteurs dont des molécules pro-inflammatoires, des protéases et des facteurs de croissance. La SASP affecte l'environnement des cellules sénescentes, en particulier au cours du vieillissement, en induisant et en modulant divers phénotypes tels que la sénescence paracrine, l'activité des cellules immunitaires et le dépôt et l'organisation de la matrice extracellulaire, qui ont un impact critique sur diverses situations physiopathologiques, notamment la fibrose et le cancer. Ici, nous découvrons un nouvel effet paracrine de la SASP : la transdifférenciation neuroendocrine (NED) de certaines cellules cancéreuses épithéliales, mise en évidence à la fois dans le sein et la prostate. Mécaniquement, cet effet est médié par des facteurs SASP dépendants de NF-κB,2+ niveaux. De manière cohérente, tamponner Ca 2+ en surexprimant la protéine tampon CALB1 inverse partiellement la NED induite par la SASP, ce qui suggère que la SASP favorise la NED par le biais d'une signalisation Ca 2+ induite par la SASP. Les analyses des ensembles de données sur le cancer du sein humain confirment que la NED survient principalement dans les tumeurs p53 WT et chez les patientes plus âgées, conformément au rôle des cellules sénescentes et de son sécrétome, car elles augmentent au cours du vieillissement. En conclusion, notre travail, révélant la NED induite par la SASP dans certaines cellules cancéreuses, ouvre la voie à de futures études visant à mieux comprendre le lien fonctionnel entre l'accumulation de cellules sénescentes au cours du vieillissement, la NED et les résultats cliniques des patients.
- Published
- 2022
- Full Text
- View/download PDF
208. Role of oxidative stress in induction of trans-differentiation of neutrophils in patients with rheumatoid arthritis
- Author
-
Aniruddha Bagchi, Parasar Ghosh, Alakendu Ghosh, and Mitali Chatterjee
- Subjects
Arthritis, Rheumatoid ,Oxidative Stress ,Neutrophils ,Cell Transdifferentiation ,Synovial Fluid ,Humans ,Vascular Cell Adhesion Molecule-1 ,General Medicine ,HLA-DR Antigens ,Intercellular Adhesion Molecule-1 ,Reactive Oxygen Species ,Biochemistry - Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder whose etiopathology involves an interplay between genetic and environmental factors, with oxidative stress being a key contributory factor. This study aimed to establish the impact, if any, of an oxidative, pro-inflammatory milieu upon trans-differentiation of neutrophils and disease progression. In the synovial fluid (SF) and peripheral blood sourced from patients with RA (
- Published
- 2022
209. Human Th1 and Th17 cells exhibit epigenetic stability at signature cytokine and transcription factor loci
- Author
-
Sarah Q. Crome, Megan K. Levings, Katherine G. MacDonald, Dixie L. Mager, C J Cohen, and Elizabeth L. Dai
- Subjects
Genetics ,Chromatin Immunoprecipitation ,Lineage (genetic) ,medicine.medical_treatment ,Gene Expression Profiling ,Immunology ,Promoter ,Biology ,Th1 Cells ,Phenotype ,Polymerase Chain Reaction ,Epigenesis, Genetic ,Cytokine ,Cell Plasticity ,Histone methylation ,Cell Transdifferentiation ,medicine ,Immunology and Allergy ,Cytokines ,Humans ,Th17 Cells ,Cell Lineage ,Epigenetics ,Transcription factor ,Transcription Factors - Abstract
The linear model of Th cell lineage commitment is being revised due to reports that mature Th cells can trans-differentiate into alternate lineages. This ability of Th cells to reprogram is thought to be regulated by epigenetic mechanisms that control expression of transcription factors characteristic of opposing lineages. It is unclear, however, to what extent this new model of Th cell plasticity holds true in human Th cell subsets that develop under physiological conditions in vivo. We isolated in vivo-differentiated human Th1 and Th17 cells, as well as intermediate Th1/17 cells, and identified distinct epigenetic signatures at cytokine (IFNG and IL17A) and transcription factor (TBX21, RORC, and RORA) loci. We also examined the phenotypic and epigenetic stability of human Th17 cells exposed to Th1-polarizing conditions and found that although they could upregulate TBX21 and IFN-γ, this occurred without loss of IL-17 or RORC expression, and resulted in cells with a Th1/17 phenotype. Similarly, Th1 cells could upregulate IL-17 upon enforced expression of RORC2, but did not lose expression of IFN-γ or TBX21. Despite alterations in expression of these signature genes, epigenetic modifications were remarkably stable aside from the acquisition of active histone methylation marks at cytokine gene promoters. The limited capacity of human Th17 and Th1 cells to undergo complete lineage conversion suggests that the bipotent Th1/17 cells may arise from Th1 and/or Th17 cells. These data also question the broad applicability of the new model of Th cell lineage plasticity to in vivo-polarized human Th cell subsets.
- Published
- 2022
- Full Text
- View/download PDF
210. Activation of α7 nicotinic acetylcholine receptor retards the development of endometriosis
- Author
-
Meihua, Hao, Xishi, Liu, and Sun-Wei, Guo
- Subjects
Mice ,Endocrinology ,alpha7 Nicotinic Acetylcholine Receptor ,Reproductive Medicine ,Cell Transdifferentiation ,Endometriosis ,Animals ,Humans ,Obstetrics and Gynecology ,Female ,Myofibroblasts ,Fibrosis ,Developmental Biology - Abstract
Background Women with endometriosis have been shown to have a reduced vagal tone as compared with controls and vagotomy promoted while vagus nerve stimulation (VNS) decelerated the progression of endometriosis in mice. Extensive research also has shown that the activation of the cholinergic anti-inflammatory pathway by VNS activates α7 nicotinic acetylcholine receptor (α7nAChR), potently reducing inflammation. Yet whether α7nAChR plays any role in endometriosis is unknown. We evaluated its expression in normal endometrium, ovarian and deep endometriotic lesions, and evaluated its role in the development of endometriosis. Methods Immunohistochemistry analyses of α7nAChR in endometriotic lesions as well as control endometrium, and quantification of tissue fibrosis by Masson trichrome staining were performed. Mouse experiments were conducted to evaluate the impact of α7nAChR activation or suppression on lesional progression and possible therapeutic effect. Finally, in vitro experiments were conducted to evaluate the effect of activation of α7nAChR on epithelial-mesenchymal transition (EMT), fibroblast-to-myofibroblast transdifferentiation (FMT), smooth muscle metaplasia (SMM) and fibrogenesis in an endometriotic epithelial cell line and primary endometriotic stromal cells derived from ovarian endometrioma tissue samples. Results Immunostaining of α7nAChR was significantly reduced in human endometriotic epithelial cells as compared with their counterpart in normal endometrium. Lesional α7nAChR staining levels correlated negatively with lesional fibrosis and the severity of dysmenorrhea. The α7nAChR agonist significantly impeded the development of endometriotic lesions in mouse models possibly through hindrance of EMT and FMT. It also demonstrated therapeutic effects in mice with induced deep endometriosis. Treatment of endometriotic epithelial and stromal cells with an α7nAChR agonist significantly abrogated platelet-induced EMT, FMT and SMM, and suppressed cellular contractility and collagen production. Conclusions α7nAChR is suppressed in endometriotic lesions, and its activation by pharmacological means can impede EMT, FMT, SMM, and fibrogenesis of endometriotic lesions. As such, α7nAChR can be rightfully viewed as a potential target for therapeutic invention. Trial registration Not applicable.
- Published
- 2022
- Full Text
- View/download PDF
211. Induced pluripotent stem cells in research and therapy of diseases: review article
- Author
-
Mohammad Reza Noori Daloii, Arash Salmaninejad, and Mina Tabrizi
- Subjects
cell transdifferentiation ,induced pluripotent stem cells (iPSCs) ,nuclear reprogramming ,treatment ,Medicine (General) ,R5-920 - Abstract
Differentiated cells can change to embryonic stem cells by reprograming. Generation of induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative and personalized medicine. iPSCs can self-renew and differentiate into many cell types. iPSC cells offer a potentially unlimited source for targeted differentiation. Through the expression of a set of transcription factors, iPSCs can be generated from different kinds of embryonic and adult cells. This technology for the first time enabled the researchers to take differentiated cells from an individual, and convert them to another cell type of interest, which is particularly to that person. When the set of master transcription factors containing OCT4, SOX2, KLF4, and MYC is expressed ectopically in somatic cells, the transcriptional network is propelled to organize itself in such a way as to maintenance a pluripotent state. Since iPSCs are similar to Embryonic Stem Cell (ESC), they can be considered as sources for modeling different diseases. iPSCs which are induced from somatic cells of patient can be useful for screening and drugs selection, and also introduce treatment via grafting the cells. Although this technology has been successful in different fields, the tumorigenesis of viral vectors during induction of reprogramming is a major challenge. Nevertheless, iPSCs are valuable for clinical applications and research. By discovery of these cells many challenges related to the safety, efficacy, and bioethics of ESCs are solved. Pluripotency is defined in two aspect of functional and molecular, by which functional regards the capacity of cell is generate three kinds of embryonic layers and germ line, and molecular aspect regards the identifying of molecules and genes that support functional features. Identification of these genes has been placed at the center of fields related to development and stem cell research. In this review, we discuss the process of generation of these cells, as well as required genes and factors for pluripotency, and also current progress in generation of iPSCs utilizing tens of reliable and new studies.
- Published
- 2014
212. Cardiomyocyte Marker Expression in Mouse Embryonic Fibroblasts by Cell-Free Cardiomyocyte Extract and Epigenetic Manipulation
- Author
-
Tahereh Talaei-Khozani, Fatemeh Heidari, Tahereh Esmaeilpour, Zahra Vojdani, Zohrah Mostafavi-Pour, and Leili Rohani
- Subjects
Cardiomyocytes ,Cell transdifferentiation ,Histone deacetylase inhibitors ,Fibroblast ,Medicine (General) ,R5-920 - Abstract
Background: The regenerative capacity of the mammalian heart is quite limited. Recent reports have focused on reprogramming mesenchymal stem cells into cardiomyocytes. We investigated whether fibroblasts could transdifferentiate into myocardium. Methods: Mouse embryonic fibroblasts were treated with Trichostatin A (TSA) and 5-Aza-2-Deoxycytidine (5-aza-dC). The treated cells were permeabilized with streptolysin O and exposed to the mouse cardiomyocyte extract and cultured for 1, 10, and 21 days. Cardiomyocyte markers were detected by immunohistochemistry. Alkaline phosphatase activity and OCT4 were also detected in cells treated by chromatin-modifying agents. Results: The cells exposed to a combination of 5-aza-dC and TSA and permeabilized in the presence of the cardiomyocyte extract showed morphological changes. The cells were unable to express cardiomyocyte markers after 24 h. Immunocytochemical assays showed a notable degree of myosin heavy chain and α-actinin expressions after 10 days. The expression of the natriuretic factor and troponin T occurred after 21 days in these cells. The cells exposed to chromatin-modifying agents also expressed cardiomyocyte markers; however, the proportion of reprogrammed cells was clearly smaller than that in the cultures exposed to 5-aza-dC , TSA, and extract. Conclusion: It seems that the fibroblasts were able to eliminate the previous epigenetic markers and form new ones according to the factors existing in the extract. Since no beating was observed, at least up to 21 days, the cells may need an appropriate extracellular matrix for their function.
- Published
- 2014
213. Conversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors
- Author
-
Ting Gong, Shan Jiang, Hitesh Chopra, Baicheng Yi, Chengfei Zhang, Ou Sha, Tian Ding, Shaohua Ge, and Waruna Lakmal Dissanayaka
- Subjects
0301 basic medicine ,Cell type ,Medicine (General) ,Angiogenesis ,Cell ,Medicine (miscellaneous) ,QD415-436 ,Chemical reprogramming ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,R5-920 ,medicine ,Dental Papilla ,Cells, Cultured ,medicine.diagnostic_test ,Chemistry ,Cell adhesion molecule ,Research ,Stem Cells ,Transdifferentiation ,Small molecules ,Endothelial Cells ,Cell Differentiation ,Endothelial differentiation ,Cell Biology ,In vitro ,Cell biology ,SCAP ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transdifferentiation ,Molecular Medicine ,Intercellular Signaling Peptides and Proteins ,Stem cell ,030217 neurology & neurosurgery - Abstract
Objectives Recently, a new strategy has been developed to directly reprogram one cell type towards another targeted cell type using small molecule compounds. Human fibroblasts have been chemically reprogrammed into neuronal cells, Schwann cells and cardiomyocyte-like cells by different small molecule combinations. This study aimed to explore whether stem cells from apical papilla (SCAP) could be reprogrammed into endothelial cells (ECs) using the same strategy. Materials and methods The expression level of endothelial-specific genes and proteins after chemical induction of SCAP was assessed by RT-PCR, western blotting, flow cytometry and immunofluorescence. The in vitro functions of SCAP-derived chemical-induced endothelial cells (SCAP-ECs) were evaluated by tube-like structure formation assay, acetylated low-density lipoprotein (ac-LDL) uptake and NO secretion detection. The proliferation and the migration ability of SCAP-ECs were evaluated by CCK-8 and Transwell assay. LPS stimulation was used to mimic the inflammatory environment in demonstrating the ability of SCAP-ECs to express adhesion molecules. The in vivo Matrigel plug angiogenesis assay was performed to assess the function of SCAP-ECs in generating vascular structures using the immune-deficient mouse model. Results SCAP-ECs expressed upregulated endothelial-specific genes and proteins; displayed endothelial transcriptional networks; exhibited the ability to form functional tubular-like structures, uptake ac-LDL and secrete NO in vitro; and contributed to generate blood vessels in vivo. The SCAP-ECs could also express adhesion molecules in the pro-inflammatory environment and have a similar migration and proliferation ability as HUVECs. Conclusions Our study demonstrates that the set of small molecules and growth factors could significantly promote endothelial transdifferentiation of SCAP, which provides a promising candidate cell source for vascular engineering and treatment of ischemic diseases.
- Published
- 2021
214. Drebrin attenuates atherosclerosis by limiting smooth muscle cell transdifferentiation
- Author
-
Francis J. Miller, Igor Nepliouev, Elizabeth R. Hauser, Tai-Qin Huang, Neil J. Freedman, Lisheng Zhang, Leigh Brian, Kamie P. Snow, Brandon M. Schickling, Jonathan A. Stiber, and Jiao-Hui Wu
- Subjects
Vascular smooth muscle ,Physiology ,Myocytes, Smooth Muscle ,Cell ,Muscle, Smooth, Vascular ,Mice ,Physiology (medical) ,medicine ,Animals ,Cells, Cultured ,Foam cell ,Mice, Knockout ,NADPH oxidase ,biology ,CD68 ,Chemistry ,Neuropeptides ,Transdifferentiation ,Original Articles ,Atherosclerosis ,musculoskeletal system ,Cell biology ,Cross-Sectional Studies ,medicine.anatomical_structure ,KLF4 ,Cell Transdifferentiation ,NADPH Oxidase 1 ,cardiovascular system ,biology.protein ,Cardiology and Cardiovascular Medicine ,tissues - Abstract
Aims The F-actin-binding protein Drebrin inhibits smooth muscle cell (SMC) migration, proliferation and pro-inflammatory signaling. Therefore, we tested the hypothesis that Drebrin constrains atherosclerosis. Methods and results SM22-Cre+/Dbnflox/flox/Ldlr-/- (SMC-Dbn-/-/Ldlr-/-) and control mice (SM22-Cre+/Ldlr-/-, Dbnflox/flox/Ldlr-/-, and Ldlr-/-) were fed a Western diet for 14-20 weeks. Brachiocephalic arteries of SMC-Dbn-/-/Ldlr-/- mice exhibited 1.5- or 1.8-fold greater cross-sectional lesion area than control mice at 14 or 20 wk, respectively. Aortic atherosclerotic lesion surface area was 1.2-fold greater in SMC-Dbn-/-/Ldlr-/- mice. SMC-Dbn-/-/Ldlr-/- lesions comprised necrotic cores that were two-fold greater in size than those of control mice. Consistent with their bigger necrotic core size, lesions in SMC-Dbn-/- arteries also showed more transdifferentiation of SMCs to macrophage-like cells: 1.5- to 2.5-fold greater, assessed with BODIPY or with CD68, respectively. In vitro data were concordant: Dbn-/- SMCs had 1.7-fold higher levels of KLF4 and transdifferentiated to macrophage-like cells more readily than Dbnflox/flox SMCs upon cholesterol loading, as evidenced by greater up-regulation of CD68 and galectin-3. Adenovirally mediated Drebrin rescue produced equivalent levels of macrophage-like transdifferentiation in Dbn-/- and Dbnflox/flox SMCs. During early atherogenesis, SMC-Dbn-/-/Ldlr-/- aortas demonstrated 1.6-fold higher levels of reactive oxygen species than control mouse aortas. The 1.8-fold higher levels of Nox1 in Dbn-/- SMCs was reduced to WT levels with KLF4 silencing. Inhibition of Nox1 chemically or with siRNA produced equivalent levels of macrophage-like transdifferentiation in Dbn-/- and Dbnflox/flox SMCs. Conclusions We conclude that SMC Drebrin limits atherosclerosis by constraining SMC Nox1 activity and SMC transdifferentiation to macrophage-like cells. Translational perspective Drebrin is abundantly expressed in vascular smooth muscle cells (SMCs) and is up-regulated in human atherosclerosis. A hallmark of atherosclerosis is the accumulation of foam cells that secrete pro-inflammatory cytokines and contribute to plaque instability. A large proportion of these foam cells in humans derive from SMCs. We found that SMC Drebrin limits atherosclerosis by reducing SMC transdifferentiation to macrophage-like foam cells in a manner dependent on Nox1 and KLF4. For this reason, strategies aimed at augmenting SMC Drebrin expression in atherosclerotic plaques may limit atherosclerosis progression and enhance plaque stability by bridling SMC-to-foam-cell transdifferentiation.
- Published
- 2021
- Full Text
- View/download PDF
215. Arecoline enhances miR-21 to promote buccal mucosal fibroblasts activation
- Author
-
Hui Wen Yang, Yi Wen Liao, Cheng Chia Yu, Chih Yuan Fang, Pei Ming Chu, Pei-Ling Hsieh, and Chuan Hang Yu
- Subjects
MicroRNA-21 ,Arecoline ,Motility ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Fibrosis ,Humans ,Medicine ,Areca ,Myofibroblast ,lcsh:R5-920 ,business.industry ,Mouth Mucosa ,General Medicine ,Fibroblasts ,Oral submucous fibrosis ,medicine.disease ,MicroRNAs ,030220 oncology & carcinogenesis ,Cell Transdifferentiation ,Cancer research ,TGF-β pathway ,030211 gastroenterology & hepatology ,business ,Carcinogenesis ,lcsh:Medicine (General) ,Transforming growth factor ,medicine.drug - Abstract
Background/purpose Oral submucous fibrosis (OSF) is an irreversible fibrosis disease and a potentially malignant disorder in the oral cavity. Various studies have shown that miR-21 was implicated in the fibrogenesis and carcinogenesis, but its functional role in the development of OSF has not been investigated. Methods The expression levels of miR-21 in arecoline-stimulated normal buccal mucosal fibroblasts (BMFs) and OSF specimens were determined by qRT-PCR. Exogenous administration of TGF-β and its inhibitor (SB431542) were utilized to examine the involvement of TGF-β signaling in miR-21 alteration. Collagen gel contraction, transwell migration, and invasion assays were used to assess the myofibroblast activities. The relationship between α-SMA and miR-21 was calculated using the Pearson correlation coefficient. Results MiR-21 expression was induced in BMFs by arecoline treatment in a dose-dependent manner. Our results showed that this upregulation was mediated by TGF-β signaling. Subsequently, we demonstrated that the administration of the miR-21 inhibitor suppressed the arecoline-induced myofibroblast characteristics, including a higher collagen gel contractility and cell motility, in normal BMFs. Furthermore, inhibition of miR-21 was sufficient to attenuate the myofibroblast features in fibrotic BMFs. Besides, we showed that the expression of miR-21 was aberrantly upregulated in the OSF tissues and there was a positive correlation between miR-21 and myofibroblast marker, α-SMA. Conclusion MiR-21 overexpression in OSF may be due to the stimulation of areca nut, which was mediated by the TGF-β pathway. Our data suggested that the repression of miR-21 was a promising direction to palliate the development and progression of OSF.
- Published
- 2021
216. Direct Reprogramming of Somatic Cells to Neurons: Pros and Cons of Chemical Approach
- Author
-
Cristiana Mollinari and Daniela Merlo
- Subjects
0301 basic medicine ,Somatic cell ,Disease ,Biology ,Biochemistry ,Regenerative medicine ,Cell therapy ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,Animals ,Humans ,Transgenes ,Epigenetics ,Neurons ,business.industry ,Gene Transfer Techniques ,Brain ,General Medicine ,Human brain ,Cellular Reprogramming ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transdifferentiation ,Commentary ,Personalized medicine ,business ,Neuroscience ,Reprogramming ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Translating successful preclinical research in neurodegenerative diseases into clinical practice has been difficult. The preclinical disease models used for testing new drugs not always appear predictive of the effects of the agents in the human disease state. Human induced pluripotent stem cells, obtained by reprogramming of adult somatic cells, represent a powerful system to study the molecular mechanisms of the disease onset and pathogenesis. However, these cells require a long time to differentiate into functional neural cells and the resetting of epigenetic information during reprogramming, might miss the information imparted by age. On the contrary, the direct conversion of somatic cells to neuronal cells is much faster and more efficient, it is safer for cell therapy and allows to preserve the signatures of donors’ age. Direct reprogramming can be induced by lineage-specific transcription factors or chemical cocktails and represents a powerful tool for modeling neurological diseases and for regenerative medicine. In this Commentary we present and discuss strength and weakness of several strategies for the direct cellular reprogramming from somatic cells to generate human brain cells which maintain age‐related features. In particular, we describe and discuss chemical strategy for cellular reprogramming as it represents a valuable tool for many applications such as aged brain modeling, drug screening and personalized medicine.
- Published
- 2021
- Full Text
- View/download PDF
217. Molecular mechanisms of transcription factor mediated cell reprogramming: conversion of liver to pancreas
- Author
-
Sebastian L. Wild and David Tosh
- Subjects
Cell type ,transdifferentiation ,Cellular differentiation ,Biology ,liver ,Biochemistry ,Regenerative medicine ,03 medical and health sciences ,0302 clinical medicine ,Directed differentiation ,Insulin-Secreting Cells ,Animals ,Humans ,pancreas ,Review Articles ,Transcription factor ,transcription factor ,030304 developmental biology ,Diabetes & Metabolic Disorders ,0303 health sciences ,Gene Expression & Regulation ,Transdifferentiation ,reprogramming ,Cellular Reprogramming ,Cell biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Cell Transdifferentiation ,Hepatocytes ,PDX1 ,Reprogramming ,Transcription Factors ,Developmental Biology - Abstract
Transdifferentiation is a type of cellular reprogramming involving the conversion of one differentiated cell type to another. This remarkable phenomenon holds enormous promise for the field of regenerative medicine. Over the last 20 years techniques used to reprogram cells to alternative identities have advanced dramatically. Cellular identity is determined by the transcriptional profile which comprises the subset of mRNAs, and therefore proteins, being expressed by a cell at a given point in time. A better understanding of the levers governing transcription factor activity benefits our ability to generate therapeutic cell types at will. One well-established example of transdifferentiation is the conversion of hepatocytes to pancreatic β-cells. This cell type conversion potentially represents a novel therapy in T1D treatment. The identification of key master regulator transcription factors (which distinguish one body part from another) during embryonic development has been central in developing transdifferentiation protocols. Pdx1 is one such example of a master regulator. Ectopic expression of vector-delivered transcription factors (particularly the triumvirate of Pdx1, Ngn3 and MafA) induces reprogramming through broad transcriptional remodelling. Increasingly, complimentary cell culture techniques, which recapitulate the developmental microenvironment, are employed to coax cells to adopt new identities by indirectly regulating transcription factor activity via intracellular signalling pathways. Both transcription factor-based reprogramming and directed differentiation approaches ultimately exploit transcription factors to influence cellular identity. Here, we explore the evolution of reprogramming and directed differentiation approaches within the context of hepatocyte to β-cell transdifferentiation focussing on how the introduction of new techniques has improved our ability to generate β-cells.
- Published
- 2021
- Full Text
- View/download PDF
218. Neurog2 directly converts astrocytes into functional neurons in midbrain and spinal cord
- Author
-
Dengyu Lu, Yuhan Shi, Xiaojing Wang, Yijie Zhang, Fei Liu, Li Sun, Fuliang Chen, Jianhua Xu, Liu Yueguang, Sanlan Li, Jiacheng Yuan, Zhiping Rao, Junlan Geng, and Sue Han
- Subjects
Cancer Research ,Neurogenesis ,Genetic Vectors ,Immunology ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Adult neurogenesis ,Article ,Midbrain ,Cellular and Molecular Neuroscience ,Glutamatergic ,Dorsal root ganglion ,Mesencephalon ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Biological neural network ,Animals ,lcsh:QH573-671 ,Cells, Cultured ,Neurons ,Oxidoreductases Acting on CH-NH Group Donors ,Glutamate Decarboxylase ,lcsh:Cytology ,Gene Transfer Techniques ,Cell Biology ,Dependovirus ,Spinal cord ,Electrophysiology ,Phenotype ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Astrocytes ,Cell Transdifferentiation ,embryonic structures ,Vesicular Glutamate Transport Protein 2 ,Neuronal development ,Ectopic expression ,Neuroscience - Abstract
Conversion of astrocytes into neurons in vivo offers an alternative therapeutic approach for neuronal loss after injury or disease. However, not only the efficiency of the conversion of astrocytes into functional neurons by single Neurog2, but also the conundrum that whether Neurog2-induced neuronal cells (Neurog2-iNs) are further functionally integrated into existing matured neural circuits remains unknown. Here, we adopted the AAV(2/8) delivery system to overexpress single factor Neurog2 into astrocytes and found that the majority of astrocytes were successfully converted into neuronal cells in multiple brain regions, including the midbrain and spinal cord. In the midbrain, Neurog2-induced neuronal cells (Neurog2-iNs) exhibit neuronal morphology, mature electrophysiological properties, glutamatergic identity (about 60%), and synapse-like configuration local circuits. In the spinal cord, astrocytes from both the intact and lesioned sources could be converted into functional neurons with ectopic expression of Neurog2 alone. Notably, further evidence from our study also proves that Neurog2-iNs in the intact spinal cord are capable of responding to diverse afferent inputs from dorsal root ganglion (DRG). Together, this study does not merely demonstrate the feasibility of Neurog2 for efficient in vivo reprogramming, it gives an indication for the Neurog2-iNs as a functional and potential factor in cell-replacement therapy.
- Published
- 2021
219. The SNAG Domain of Insm1 Regulates Pancreatic Endocrine Cell Differentiation and Represses β- to δ-Cell Transdifferentiation
- Author
-
Jingshen Zhuang, Weihua Tao, Ulrich Koestner, Shiqi Jia, Cunchuan Wang, Huimin Li, Yahui Mao, Xuehua Liang, Feng Deng, Luis R. Hernandez-Miranda, Hualin Duan, and Yiqiu Wei
- Subjects
0301 basic medicine ,Enteroendocrine Cells ,Endocrinology, Diabetes and Metabolism ,Mutant ,Hematopoietically expressed homeobox ,030209 endocrinology & metabolism ,Enteroendocrine cell ,medicine.disease_cause ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin-Secreting Cells ,Internal Medicine ,medicine ,Animals ,Transcription factor ,Homeodomain Proteins ,Zinc finger ,Mutation ,biology ,Transdifferentiation ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell biology ,Repressor Proteins ,030104 developmental biology ,Cell Transdifferentiation ,biology.gene ,Transcription Factors - Abstract
The allocation and specification of pancreatic endocrine lineages are tightly regulated by transcription factors. Disturbances in differentiation of these lineages contribute to the development of various metabolic diseases, including diabetes. The insulinoma-associated protein 1 (Insm1), which encodes a protein containing one SNAG domain and five zinc fingers, plays essential roles in pancreatic endocrine cell differentiation and in mature β-cell function. In the current study, we compared the differentiation of pancreatic endocrine cells between Insm1 null and Insm1 SNAG domain mutants (Insm1delSNAG) to explore the specific function of the SNAG domain of Insm1. We show that the δ-cell number is increased in Insm1delSNAG but not in Insm1 null mutants as compared with the control mice. We also show a less severe reduction of the β-cell number in Insm1delSNAG as that in Insm1 null mutants. In addition, similar deficits are observed in α-, PP, and ε-cells in Insm1delSNAG and Insm1 null mutants. We further identified that the increased δ-cell number is due to β- to δ-cell transdifferentiation. Mechanistically, the SNAG domain of Insm1 interacts with Lsd1, the demethylase of H3K4me1/2. Mutation in the SNAG domain of Insm1 results in impaired recruitment of Lsd1 and increased H3K4me1/2 levels at hematopoietically expressed homeobox (Hhex) loci that are bound by Insm1, thereby promoting the transcriptional activity of the δ-cell–specific gene Hhex. Our study has identified a novel function of the SNAG domain of Insm1 in the regulation of pancreatic endocrine cell differentiation, particularly in the repression of β- to δ-cell transdifferentiation.
- Published
- 2021
- Full Text
- View/download PDF
220. Therapy-Induced Transdifferentiation Promotes Glioma Growth Independent of EGFR Signaling
- Author
-
Jun Yao, Inah Hwang, Hongwu Zheng, J. Li, Qianghu Wang, Lingxiang Wu, Baoli Hu, Dongqing Cao, Ja Young Jang, Haoqiang Ying, Yu Yao, Hwanhee Oh, and Jihye Paik
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Datasets as Topic ,Apoptosis ,Transcriptome ,Mice ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,RNA-Seq ,Epidermal growth factor receptor ,Mice, Knockout ,education.field_of_study ,biology ,Brain Neoplasms ,Transdifferentiation ,Glioma ,Prognosis ,Progression-Free Survival ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Female ,Signal Transduction ,Slug ,Population ,Transforming Growth Factor beta1 ,Erlotinib Hydrochloride ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,education ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Homeodomain Proteins ,Mesenchymal stem cell ,YAP-Signaling Proteins ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cell Transdifferentiation ,Cancer research ,biology.protein ,Neoplasm Recurrence, Local ,Transcription Factors - Abstract
EGFR is frequently amplified, mutated, and overexpressed in malignant gliomas. Yet the EGFR-targeted therapies have thus far produced only marginal clinical responses, and the underlying mechanism remains poorly understood. Using an inducible oncogenic EGFR-driven glioma mouse model system, our current study reveals that a small population of glioma cells can evade therapy-initiated apoptosis and potentiate relapse development by adopting a mesenchymal-like phenotypic state that no longer depends on oncogenic EGFR signaling. Transcriptome analyses of proximal and distal treatment responses identified TGFβ/YAP/Slug signaling cascade activation as a major regulatory mechanism that promotes therapy-induced glioma mesenchymal lineage transdifferentiation. Following anti-EGFR treatment, TGFβ secreted from stressed glioma cells acted to promote YAP nuclear translocation that stimulated upregulation of the pro-mesenchymal transcriptional factor SLUG and subsequent glioma lineage transdifferentiation toward a stable therapy-refractory state. Blockade of this adaptive response through suppression of TGFβ-mediated YAP activation significantly delayed anti-EGFR relapse and prolonged animal survival. Together, our findings shed new insight into EGFR-targeted therapy resistance and suggest that combinatorial therapies of targeting both EGFR and mechanisms underlying glioma lineage transdifferentiation could ultimately lead to deeper and more durable responses. Significance: This study demonstrates that molecular reprogramming and lineage transdifferentiation underlie anti-EGFR therapy resistance and are clinically relevant to the development of new combinatorial targeting strategies against malignant gliomas with aberrant EGFR signaling.
- Published
- 2021
- Full Text
- View/download PDF
221. TMT-based quantitative proteome profiles reveal the memory function of a whole heart decellularized matrix for neural stem cell trans-differentiation into the cardiac lineage
- Author
-
Zhongbao Gao, Baijun Liu, Chunlan Wang, Yuan Shen, Wei Liu, Qi Yin, Xiaoning Yang, Changyong Wang, Xiao Zhang, and Jin Zhou
- Subjects
0303 health sciences ,Decellularization ,Proteome ,Tissue Engineering ,Tissue Scaffolds ,biology ,Quantitative proteomics ,Integrin ,Biomedical Engineering ,02 engineering and technology ,021001 nanoscience & nanotechnology ,Neural stem cell ,Extracellular Matrix ,Cell biology ,Extracellular matrix ,03 medical and health sciences ,Neural Stem Cells ,Tissue engineering ,Cell Transdifferentiation ,biology.protein ,General Materials Science ,Stem cell ,0210 nano-technology ,030304 developmental biology - Abstract
Whole organ or tissue decellularized matrices are a promising scaffold for tissue engineering because they maintain the specific memory of the original organ or tissue. A whole organ or tissue decellularized matrix contains extracellular matrix (ECM) components, and exhibits ultrastructural and mechanical properties, which could significantly regulate the fate of stem cells. To better understand the memory function of whole organ decellularized matrices, we constructed a heart decellularized matrix and seeded cross-embryonic layer stem cells - neural stem cells (NSCs) to repopulate the matrix, engineering cardiac tissue, in which a large number of NSCs differentiated into the neural lineage, but besides that, NSCs showed an obvious tendency of trans-differentiating into cardiac lineage cells. The results demonstrated that the whole heart decellularized microenvironment possesses memory function. To reveal the underlying mechanism, TMT-based quantitative proteomics analysis was used to identify the differently expressed proteins in the whole heart decellularized matrix compared with a brain decellularized matrix. 937 of the proteins changed over 1.5 fold, with 573 of the proteins downregulated and 374 of the proteins upregulated, among which integrin ligands in the ECM serve as key signals in regulating NSC fate. The findings here provide a novel insight into the memory function of tissue-specific microenvironments and pave the way for the therapeutic application of personalized tissues.
- Published
- 2021
- Full Text
- View/download PDF
222. Myocyte-specific enhancer factor 2c triggers transdifferentiation of adipose tissue-derived stromal cells into spontaneously beating cardiomyocyte-like cells
- Author
-
Masa-aki Kawashiri, Soichiro Usui, Shinichiro Takashima, Masayuki Takamura, Kenji Sakata, Kenshi Hayashi, Shihe Cui, Oto Inoue, Kosei Yamaguchi, Yusuke Takeda, Chiaki Goten, and Yoshio Sakai
- Subjects
0301 basic medicine ,Male ,Pluripotent Stem Cells ,Stromal cell ,Science ,Adipose tissue ,Stem-cell differentiation ,030204 cardiovascular system & hematology ,Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Animals ,MEF2C ,Myocytes, Cardiac ,Progenitor cell ,Induced pluripotent stem cell ,Cells, Cultured ,health care economics and organizations ,Cell Proliferation ,Adult stem cells ,Multidisciplinary ,MEF2 Transcription Factors ,Regeneration (biology) ,Transdifferentiation ,Reprogramming ,Cell Differentiation ,Mesenchymal Stem Cells ,Stromal vascular fraction ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Adipose Tissue ,Cell Transdifferentiation ,cardiovascular system ,Medicine ,Female ,Stromal Cells - Abstract
Cardiomyocyte regeneration is limited in adults. The adipose tissue-derived stromal vascular fraction (Ad-SVF) contains pluripotent stem cells that rarely transdifferentiate into spontaneously beating cardiomyocyte-like cells (beating CMs). However, the characteristics of beating CMs and the factors that regulate the differentiation of Ad-SVF toward the cardiac lineage are unknown. We developed a simple culture protocol under which the adult murine inguinal Ad-SVF reproducibly transdifferentiates into beating CMs without induction. The beating CMs showed the striated ventricular phenotype of cardiomyocytes and synchronised oscillation of the intracellular calcium concentration among cells on day 28 of Ad-SVF primary culture. We also identified beating CM-fated progenitors (CFPs) and performed single-cell transcriptome analysis of these CFPs. Among 491 transcription factors that were differentially expressed (≥ 1.75-fold) in CFPs and the beating CMs, myocyte-specific enhancer 2c (Mef2c) was key. Transduction of Ad-SVF cells with Mef2c using a lentiviral vector yielded CFPs and beating CMs with ~ tenfold higher cardiac troponin T expression, which was abolished by silencing of Mef2c. Thus, we identified the master gene required for transdifferentiation of Ad-SVF into beating CMs. These findings will facilitate the development of novel cardiac regeneration therapies based on gene-modified, cardiac lineage-directed Ad-SVF cells.
- Published
- 2021
223. Brown and beige adipose tissue: a novel therapeutic strategy for obesity and type 2 diabetes mellitus
- Author
-
Haifeng Du, Quantao Ma, Jingkang Wang, Chen Wang, Hongyu Dai, Yaqi Li, Pengfei Li, Yinglan Lv, Bao-Sheng Zhao, Yongcheng An, Lu Shi, Long Cheng, Huimin Li, and Yuhui Duan
- Subjects
medicine.medical_specialty ,obesity ,Histology ,Physiology ,beige adipose tissue ,type 2 diabetes mellitus ,Adipose Tissue, White ,Adipocytes, White ,Adipose tissue ,White adipose tissue ,Review ,Biology ,Brown adipose tissue ,Diseases of the endocrine glands. Clinical endocrinology ,Insulin resistance ,white adipose tissue ,Adipose Tissue, Brown ,Internal medicine ,medicine ,Browning ,QP1-981 ,browning of white adipose tissue ,Humans ,Adipocytes, Beige ,QH573-671 ,Transdifferentiation ,Type 2 Diabetes Mellitus ,nutritional and metabolic diseases ,food and beverages ,Thermogenesis ,Cell Biology ,Adipose Tissue, Beige ,medicine.disease ,RC648-665 ,Obesity ,Endocrinology ,medicine.anatomical_structure ,Adipocytes, Brown ,Adipose Tissue ,Diabetes Mellitus, Type 2 ,Cell Transdifferentiation ,Insulin Resistance ,Cytology ,Energy Metabolism - Abstract
Mammalian adipose tissue can be divided into two major types, namely, white adipose tissue (WAT) and brown adipose tissue (BAT). According to classical view, the main function of WAT is to store excess energy in the form of triglycerides, while BAT is a thermogenic tissue that acts a pivotal part in maintaining the core body temperature. White adipocytes display high plasticity and can transdifferentiate into beige adipocytes which have many similar morphological and functional properties with brown adipocytes under the stimulations of exercise, cold exposure and other factors. This phenomenon is also known as ‘browning of WAT’. In addition to transdifferentiation, beige adipocytes can also come from de novo differentiation from tissue-resident progenitors. Activating BAT and inducing browning of WAT can accelerate the intake of glycolipids and reduce the insulin secretion requirement, which may be a new strategy to improve glycolipids metabolism and insulin resistance of obese and type 2 diabetes mellitus (T2DM) patients. This review mainly discusses the significance of brown and beige adipose tissues in the treatment of obesity and T2DM, and focuses on the effect of the browning agent on obesity and T2DM, which provides a brand-new theoretical reference for the prevention and treatment of obesity and T2DM.
- Published
- 2021
224. There and back again: The mechanisms of differentiation and transdifferentiation in Drosophila blood cells
- Author
-
Viktor Honti, Erika Gábor, and Gábor Csordás
- Subjects
Cell type ,Hemocytes ,Cellular differentiation ,Transcriptome ,Blood cell ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Molecular Biology ,030304 developmental biology ,Phagocytes ,0303 health sciences ,Innate immune system ,biology ,Transdifferentiation ,Cell Differentiation ,Cell Biology ,biology.organism_classification ,Hematopoiesis ,Cell biology ,Drosophila melanogaster ,medicine.anatomical_structure ,Cell Transdifferentiation ,Stem cell ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Transdifferentiation is a conversion of an already differentiated cell type into another cell type without the involvement of stem cells. This transition is well described in the case of vertebrate immune cells, as well as in Drosophila melanogaster, which therefore serves as a suitable model to study the process in detail. In the Drosophila larva, the latest single-cell sequencing methods enabled the clusterization of the phagocytic blood cells, the plasmatocytes, which are capable of transdifferentiation into encapsulating cells, the lamellocytes. Here we summarize the available data of the past years on the plasmatocyte-lamellocyte transition, and make an attempt to harmonize them with transcriptome-based blood cell clustering to better understand the underlying mechanisms of transdifferentiation in Drosophila, and in general.
- Published
- 2021
- Full Text
- View/download PDF
225. Metabolic Adaptations to Pregnancy in Healthy and Gestational Diabetic Pregnancies: The Pancreas - Placenta Axis
- Author
-
David J. Hill and Sandra K Szlapinski
- Subjects
Blood Glucose ,0301 basic medicine ,medicine.medical_specialty ,Placenta ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Risk Assessment ,Obesity, Maternal ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Pregnancy ,Risk Factors ,Insulin-Secreting Cells ,Internal medicine ,Animals ,Humans ,Medicine ,Pancreas ,Cell Proliferation ,Pharmacology ,Fetus ,business.industry ,Insulin ,Pregnancy Outcome ,medicine.disease ,Adaptation, Physiological ,3. Good health ,Gestational diabetes ,Diabetes, Gestational ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cell Transdifferentiation ,Gestation ,Female ,Inflammation Mediators ,Energy Metabolism ,Cardiology and Cardiovascular Medicine ,business ,Hormone - Abstract
Normal pregnancy is associated with increased insulin resistance as a metabolic adaptation to the nutritional demands of the placenta and fetus, and this is amplified in obese mothers. Insulin resistance is normally compensated for by an adaptive increase in pancreatic β-cell mass together with enhanced glucose-stimulated insulin release. Placentally-derived hormones and growth factors are central to the altered pancreatic morphology and function. A failure of β-cells to undergo adaptive change after the first trimester has been linked with gestational diabetes. In the pregnant mouse, an increase in β-cell replication contributes to a 2-3-fold increase in mass peaking in late gestation, depending on the proliferation of existing β-cells, the differentiation of resident progenitor β-cells, or islet cell transdifferentiation. Using mouse models and human studies placenta- and islet of Langerhans-derived molecules have been identified that are likely to contribute to the metabolic adaptations to pregnancy and whose physiology is altered in the obese, glucose-intolerant mother. Maternal obesity during pregnancy can create a pro-inflammatory environment that can disrupt the response of the β-cells to the endocrine signals of pregnancy and limit the adaptive changes in β-cell mass and function, resulting in an increased risk of gestational diabetes.
- Published
- 2020
- Full Text
- View/download PDF
226. Combination of tumor necrosis factor‐α and epidermal growth factor induces the adrenergic‐to‐mesenchymal transdifferentiation in SH‐SY5Y neuroblastoma cells
- Author
-
Nobuhiro Nishio, Shoma Tsubota, Kenji Kadomatsu, Yoshiyuki Takahashi, and Yue Huang
- Subjects
Adrenergic Neurons ,0301 basic medicine ,Cancer Research ,Cell type ,animal structures ,intratumor heterogeneity ,Biology ,Neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,Cell, Molecular, and Stem Cell Biology ,Epidermal growth factor ,Cell Line, Tumor ,medicine ,Humans ,Transcription factor ,Epidermal Growth Factor ,Tumor Necrosis Factor-alpha ,Transdifferentiation ,Mesenchymal stem cell ,Original Articles ,General Medicine ,tumor necrosis factor‐alpha ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cell Transdifferentiation ,Cancer research ,Original Article ,Tumor necrosis factor alpha - Abstract
Neuroblastoma, a type of cancer that is common in children, is composed of two genetically clonal but epigenetically distinct cell types: mesenchymal (MES) and adrenergic (ADRN) types, controlled by super‐enhancer‐associated lineage‐specific transcription factor networks. Mesenchymal‐type cells are more migratory, resistant to chemotherapy, and prevalent in relapse tumors. Importantly, both cell types spontaneously transdifferentiate into one another, and this interconversion can be induced by genetic manipulations. However, the mechanisms of their spontaneous transdifferentiation and extracellular factors inducing this phenomenon have not yet been elucidated. Using a unique approach involving gene set enrichment analysis, we selected six ADRN and 10 MES candidate factors, possibly inducing ADRN and MES phenotypes, respectively. Treatment with a combination of 10 MES factors clearly induced the MES gene expression profile in ADRN‐type SH‐SY5Y neuroblastoma cells. Considering the effects on gene expression profile, migration ability, and chemoresistance, a combination of tumor necrosis factor alpha (TNF‐α) and epidermal growth factor (EGF) was sufficient to synergistically induce the ADRN‐to‐MES transdifferentiation in SH‐SY5Y cells. In addition, human neuroblastoma cohort analysis revealed that the expression of TNF and EGF receptors was strongly associated with MES gene expression signatures, supporting their important roles in transdifferentiation in vivo. Collectively, we propose a mechanism of neuroblastoma transdifferentiation induced by extracellular growth factors, which can be controlled in clinical situations, providing a new therapeutic possibility., Neuroblastoma is composed of two epigenetically controlled cell types, mesenchymal (MES) and adrenergic (ADRN) types, which spontaneously transdifferentiate into one another. We found a synergistic combination of tumor necrosis factor and epidermal growth factor that induced the transdifferentiation from ADRN to MES state in ADRN‐type SH‐SY5Y neuroblastoma cells.
- Published
- 2020
- Full Text
- View/download PDF
227. A long noncoding RNA, LncMyoD, modulates chromatin accessibility to regulate muscle stem cell myogenic lineage progression
- Author
-
Tom H. Cheung, Wai-Kin So, Christopher B. Preusch, Kangning Lin, Ran Yi, Zhenguo Wu, Shaoyuan Luan, Joyce W Wong, and Anqi Dong
- Subjects
Male ,Satellite Cells, Skeletal Muscle ,chromatin modulation ,Biology ,Cell fate determination ,MyoD ,Muscle Development ,Myoblasts ,lncRNA ,Animals ,Cell Lineage ,Epigenetics ,MyoD Protein ,Multidisciplinary ,lineage progression ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Biological Sciences ,Fibroblasts ,Chromatin ,Cell biology ,stem cell ,Mice, Inbred C57BL ,Regulatory sequence ,Myogenic regulatory factors ,Cell Transdifferentiation ,Female ,RNA, Long Noncoding ,Stem cell ,Reprogramming ,Developmental Biology - Abstract
Significance Epigenetic regulations control the accessibility of transcription factors to their target regions. Modulation of chromatin accessibility determines which transcripts to be expressed and therefore, defines cell identity. Chromatin modulation during cell fate determination involves a complex regulatory network, yet the comprehensive view remains to be explored. Here, we provide a global view of chromatin accessibility during muscle stem cell activation. We identified a long noncoding RNA (lncRNA), LncMyoD, which regulates lineage determination and progression through modulating chromatin accessibility. Functional analysis showed that loss of LncMyoD strongly impairs reprogramming of fibroblasts into myogenic lineage and causes defects in muscle stem cell differentiation. Our findings provide an epigenetic mechanism for the regulation of muscle stem cell myogenic lineage progression by an lncRNA., Epigenetics regulation plays a critical role in determining cell identity by controlling the accessibility of lineage-specific regulatory regions. In muscle stem cells, epigenetic mechanisms of how chromatin accessibility is modulated during cell fate determination are not fully understood. Here, we identified a long noncoding RNA, LncMyoD, that functions as a chromatin modulator for myogenic lineage determination and progression. The depletion of LncMyoD in muscle stem cells led to the down-regulation of myogenic genes and defects in myogenic differentiation. LncMyoD exclusively binds with MyoD and not with other myogenic regulatory factors and promotes transactivation of target genes. The mechanistic study revealed that loss of LncMyoD prevents the establishment of a permissive chromatin environment at myogenic E-box–containing regions, therefore restricting the binding of MyoD. Furthermore, the depletion of LncMyoD strongly impairs the reprogramming of fibroblasts into the myogenic lineage. Taken together, our study shows that LncMyoD associates with MyoD and promotes myogenic gene expression through modulating MyoD accessibility to chromatin, thereby regulating myogenic lineage determination and progression.
- Published
- 2020
228. LncRNA HOTAIR promotes endometrial fibrosis by activating TGF-β1/Smad pathway
- Author
-
Yinmei Dai, Lingge Jin, Dan Lu, Liang Huang, Jing Chen, Juhong Liu, Ziwen Jiang, Yudi Zhang, Aihong Duan, Zhaohui Liu, and Jianhong Wu
- Subjects
Adult ,0301 basic medicine ,Stromal cell ,Primary Cell Culture ,Biophysics ,Apoptosis ,Tissue Adhesions ,Smad2 Protein ,SMAD ,Biochemistry ,Collagen Type I ,Transforming Growth Factor beta1 ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,medicine ,Humans ,Gene silencing ,Smad3 Protein ,Cell Proliferation ,Uterine Diseases ,biology ,Chemistry ,HOTAIR ,General Medicine ,Transforming growth factor beta ,medicine.disease ,Actins ,Up-Regulation ,Antisense RNA ,Collagen Type I, alpha 1 Chain ,030104 developmental biology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Cell Transdifferentiation ,embryonic structures ,Cancer research ,biology.protein ,Female ,RNA, Long Noncoding ,Stromal Cells ,Signal Transduction ,Transforming growth factor - Abstract
Homeobox transcript antisense RNA (HOTAIR) is a long non-coding RNA associated with a number of fibrosis-related diseases. The aim of this study was to investigate the specific role of HOTAIR in the development of endometrial fibrosis and to identify the molecular mechanisms underlying this process. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to determine the expression levels of HOTAIR in samples of intrauterine adhesion (IUA) tissue and in endometrial stromal cells (ESCs) that had been treated with transforming growth factor beta 1 (TGF-β1). Additionally, we transfected ESCs with either overexpression plasmid (pcDNA-HOTAIR) or silencing construct (si-HOTAIR) and then treated these cells with TGF-β1. We then performed RT-qPCR and western blot analysis, along with cell proliferation and apoptosis assays, to investigate the effects of HOTAIR on the transdifferentiation of ESCs into myofibroblasts. The results showed that the expression levels of HOTAIR were significantly elevated in IUA tissue and in ESCs that had been treated with TGF-β1. The overexpression of HOTAIR had a pro-fibrotic effect on ESCs, while the silencing of HOTAIR exerted an anti-fibrotic effect. Most importantly, the protein expression levels of p-Smad2 and p-Smad3 were significantly upregulated in TGF-β1-treated ESCs transfected with pcDNA-HOTAIR and were downregulated after transfection with si-HOTAIR constructs. These data indicate that HOTAIR promotes endometrial fibrosis by activating the TGF-β1/Smad signaling pathway, suggesting that the inhibition of HOTAIR may represent a promising therapeutic option for suppressing endometrial fibrosis.
- Published
- 2020
- Full Text
- View/download PDF
229. In Vitro Differentiation of Human Amniotic Epithelial Cells into Hepatocyte-like Cells
- Author
-
Marcin Michalik, Patrycja Wieczorek, and Piotr Czekaj
- Subjects
Epidermal Growth Factor ,amniotic epithelial cells ,stem cells ,differentiation ,hepatocytes ,EGF ,HGF ,Cell Transdifferentiation ,Induced Pluripotent Stem Cells ,Hepatocytes ,Humans ,Cellular Reprogramming Techniques ,Epithelial Cells ,General Medicine ,Amnion ,Cells, Cultured - Abstract
Human amniotic epithelial cells (hAECs) represent an interesting clinical alternative to human embryonic (hESCs) and induced pluripotent (hiPSCs) stem cells in regenerative medicine. The potential of hAECs can be enhanced ex vivo by their partial pre-differentiation. The aim of this study was to evaluate the effectiveness of 18-day differentiation of hAECs into endodermal cells, hepatic precursor cells, and cells showing functional features of hepatocytes using culture media supplemented with high (100 ng/mL) concentrations of EGF or HGF. The cells obtained after differentiation showed changes in morphology and increased expression of AFP, ALB, CYP3A4, CYP3A7, and GSTP1 genes. HGF was more effective than EGF in increasing the expression of liver-specific genes in hAECs. However, EGF stimulated the differentiation process more efficiently and yielded more hepatocyte-like cells capable of synthesizing α-fetoprotein during differentiation. Additionally, after 18 days, GST transferases, albumin, and CYP P450s, which proved their partial functionality, were expressed. In summary, HGF and EGF at a dose of 100 ng/mL can be successfully used to obtain hepatocyte-like cells between days 7 and 18 of hAEC differentiation. However, the effectiveness of this process is lower compared with hiPSC differentiation; therefore, optimization of the composition of the medium requires further research.
- Published
- 2022
230. Generation of specialized blood vessels via lymphatic transdifferentiation
- Author
-
Rudra N. Das, Yaara Tevet, Stav Safriel, Yanchao Han, Noga Moshe, Giuseppina Lambiase, Ivan Bassi, Julian Nicenboim, Matthias Brückner, Dana Hirsch, Raya Eilam-Altstadter, Wiebke Herzog, Roi Avraham, Kenneth D. Poss, and Karina Yaniv
- Subjects
Multidisciplinary ,Cell Transdifferentiation ,Animal Fins ,Animals ,Blood Vessels ,Endothelial Cells ,Cell Lineage ,Article ,Zebrafish ,Lymphatic Vessels - Abstract
The lineage and developmental trajectory of a cell are key determinants of cellular identity. In the vascular system, endothelial cells (ECs) of blood and lymphatic vessels (LVs) differentiate and specialize to cater the unique physiological demands of each organ(1,2). While LVs were shown to derive from multiple cellular origins, lymphatic ECs (LECs) are not known to generate other cell-types(3,4). Here, we use recurrent imaging and lineage-tracing of ECs in zebrafish anal fins (AF), from early development through adulthood, to uncover an unexpected mechanism of specialized blood vessel formation through transdifferentiation of LECs. Moreover, we demonstrate that deriving AF vessels from lymphatic vs. blood ECs results in functional differences in the adult organism, uncovering a link between cell ontogeny and functionality. We further use scRNA-seq to characterize the different cellular populations and transition states involved in the transdifferentiation process. Finally, we show that akin to normal development, the vasculature is re-derived from lymphatics during AF regeneration, demonstrating that LECs in adult fish retain both potency and plasticity for generating blood ECs. Overall, our work highlights a new innate mechanism of blood vessel formation through LEC transdifferentiation, and provides in vivo evidence for a link between cell ontogeny and functionality in ECs.
- Published
- 2022
231. TGFβ1 induces myofibroblast transdifferentiation via increasing Smad-mediated RhoGDI-RhoGTPase signaling
- Author
-
Lian Tang, Panfeng Feng, Yan Qi, Lei Huang, Xiuying Liang, and Xia Chen
- Subjects
rho Guanine Nucleotide Dissociation Inhibitor alpha ,Physiology ,Cell Transdifferentiation ,Biophysics ,Humans ,rho-Specific Guanine Nucleotide Dissociation Inhibitors ,General Medicine ,Myofibroblasts ,Signal Transduction - Abstract
This study serves to investigate the effects of the Smad pathway on TGFβ1-mediated RhoGDI expression and its binding to RhoGTPases in myofibroblast transdifferentiation. Myofibroblast transdifferentiation was induced by TGFβ1 in vitro. Cells were pretreated with different siRNAs or inhibitors. Myofibroblast transdifferentiation was detected by immunohistochemistry. Immunofluorescence was used to observe the nuclear translocation of Smad4, and PSR (Picrositius Red) staining was used to measure collagen concentration. TGFβ1 induced the phosphorylation of Smad2/3 and the nuclear translocation of Smad4 in human aortic adventitial fibroblasts (HAAFs). Furthermore, TGFβ1 increased the expression of RhoGDI and its binding to RhoGTPases. Nevertheless, inhibition of Smad2/3 phosphorylation decreased TGFβ1-induced RhoGDI1/2 expressions and RhoGDI2-RhoGTPases interactions. These data suggested that the inhibition of Smad phosphorylation attenuates myofibroblast transdifferentiation by inhibiting TGFβ1-induced RhoGDI1/2 expressions and RhoGDI-RhoGTPases signaling.
- Published
- 2022
232. Pancreatic Transdifferentiation Using β-Cell Transcription Factors for Type 1 Diabetes Treatment
- Author
-
Alexandra L. G. Mahoney, Najah T. Nassif, Bronwyn A. O’Brien, and Ann M. Simpson
- Subjects
Diabetes Mellitus, Type 1 ,Insulin-Secreting Cells ,Cell Transdifferentiation ,Humans ,Insulin ,Cellular Reprogramming Techniques ,General Medicine ,Transcription Factors - Abstract
Type 1 diabetes is a chronic illness in which the native beta (β)-cell population responsible for insulin release has been the subject of autoimmune destruction. This condition requires patients to frequently measure their blood glucose concentration and administer multiple daily exogenous insulin injections accordingly. Current treatments fail to effectively treat the disease without significant side effects, and this has led to the exploration of different approaches for its treatment. Gene therapy and the use of viral vectors has been explored extensively and has been successful in treating a range of diseases. The use of viral vectors to deliver β-cell transcription factors has been researched in the context of type 1 diabetes to induce the pancreatic transdifferentiation of cells to replace the β-cell population destroyed in patients. Studies have used various combinations of pancreatic and β-cell transcription factors in order to induce pancreatic transdifferentiation and have achieved varying levels of success. This review will outline why pancreatic transcription factors have been utilised and how their application can allow the development of insulin-producing cells from non β-cells and potentially act as a cure for type 1 diabetes.
- Published
- 2022
233. Plaque Evaluation by Ultrasound and Transcriptomics Reveals BCLAF1 as a Regulator of Smooth Muscle Cell Lipid Transdifferentiation in Atherosclerosis
- Author
-
Urszula Rykaczewska, Quanyi Zhao, Peter Saliba-Gustafsson, Mariette Lengquist, Malin Kronqvist, Otto Bergman, Zhiqiang Huang, Kent Lund, Katarina Waden, Zara Pons Vila, Kenneth Caidahl, Josefin Skogsberg, Vladana Vukojevic, Jan H.N. Lindeman, Joy Roy, Göran K. Hansson, Eckardt Treuter, Nicholas J. Leeper, Per Eriksson, Ewa Ehrenborg, Anton Razuvaev, Ulf Hedin, and Ljubica Matic
- Subjects
endarterectomy ,Tumor Suppressor Proteins ,Myocytes, Smooth Muscle ,Lipids ,Plaque, Atherosclerotic ,Repressor Proteins ,carotid ,Mice ,Proto-Oncogene Proteins c-bcl-2 ,Cell Transdifferentiation ,Animals ,atherosclerosis ,Cardiology and Cardiovascular Medicine ,humans ,mitogens ,transcriptome ,Ultrasonography - Abstract
Background: Understanding the processes behind carotid plaque instability is necessary to develop methods for identification of patients and lesions with stroke risk. Here, we investigated molecular signatures in human plaques stratified by echogenicity as assessed by duplex ultrasound. Methods: Lesion echogenicity was correlated to microarray gene expression profiles from carotid endarterectomies (n=96). The findings were extended into studies of human and mouse atherosclerotic lesions in situ, followed by functional investigations in vitro in human carotid smooth muscle cells (SMCs). Results: Pathway analyses highlighted muscle differentiation, iron homeostasis, calcification, matrix organization, cell survival balance, and BCLAF1 (BCL2 [B-cell lymphoma 2]-associated transcription factor 1) as the most significant signatures. BCLAF1 was downregulated in echolucent plaques, positively correlated to proliferation and negatively to apoptosis. By immunohistochemistry, BCLAF1 was found in normal medial SMCs. It was repressed early during atherogenesis but reappeared in CD68+ cells in advanced plaques and interacted with BCL2 by proximity ligation assay. In cultured SMCs, BCLAF1 was induced by differentiation factors and mitogens and suppressed by macrophage-conditioned medium. BCLAF1 silencing led to downregulation of BCL2 and SMC markers, reduced proliferation, and increased apoptosis. Transdifferentiation of SMCs by oxLDL (oxidized low-denisty lipoprotein) was accompanied by upregulation of BCLAF1, CD36, and CD68, while oxLDL exposure with BCLAF1 silencing preserved MYH (myosin heavy chain) 11 expression and prevented transdifferentiation. BCLAF1 was associated with expression of cell differentiation, contractility, viability, and inflammatory genes, as well as the scavenger receptors CD36 and CD68 . BCLAF1 expression in CD68+/BCL2+ cells of SMC origin was verified in plaques from MYH11 lineage-tracing atherosclerotic mice. Moreover, BCLAF1 downregulation associated with vulnerability parameters and cardiovascular risk in patients with carotid atherosclerosis. Conclusions: Plaque echogenicity correlated with enrichment of distinct molecular pathways and identified BCLAF1 , previously not described in atherosclerosis, as the most significant gene. Functionally, BCLAF1 seems necessary for survival and transdifferentiation of SMCs into a macrophage-like phenotype. The role of BCLAF1 in plaque vulnerability should be further evaluated.
- Published
- 2022
234. The role of DNA demethylation in liver to pancreas transdifferentiation
- Author
-
Adi Har-Zahav, Daniela Lixandru, David Cheishvili, Ioan Valentin Matei, Ioana Raluca Florea, Veronica Madalina Aspritoiu, Inna Blus-Kadosh, Irit Meivar-Levy, Andreea Madalina Serban, Irinel Popescu, Moshe Szyf, Sarah Ferber, and Simona Olimpia Dima
- Subjects
DNA Demethylation ,Liver ,Cell Transdifferentiation ,Insulins ,Molecular Medicine ,Medicine (miscellaneous) ,Cell Biology ,DNA ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Pancreas ,Chromatin ,Transcription Factors - Abstract
Background Insulin producing cells generated by liver cell transdifferentiation, could serve as an attractive source for regenerative medicine. The present study assesses the relationship between DNA methylation pTFs induced liver to pancreas transdifferentiation. Results The transdifferentiation process is associated with DNA demethylation, mainly at gene regulatory sites, and with increased expression of these genes. Active inhibition of DNA methylation promotes the pancreatic transcription factor-induced transdifferentiation process, supporting a causal role for DNA demethylation in this process. Conclusions Transdifferentiation is associated with global DNA hypomethylation, and with increased expression of specific demethylated genes. A combination of epigenetic modulators may be used to increase chromatin accessibility of the pancreatic transcription factors, thus promoting the efficiency of the developmental process.
- Published
- 2022
235. Valproic Acid Initiates Transdifferentiation of the Human Ductal Adenocarcinoma Cell-line Panc-1 Into α-Like Cells
- Author
-
Sebastian Friedrich Petry, Naga Deepa Kandula, Stefan Günther, Christian Helker, Undraga Schagdarsurengin, and Thomas Linn
- Subjects
Histones ,Histone Deacetylase Inhibitors ,Endocrinology ,Endocrinology, Diabetes and Metabolism ,Valproic Acid ,Cell Transdifferentiation ,Internal Medicine ,Insulins ,Humans ,General Medicine ,Adenocarcinoma ,Epigenesis, Genetic ,Transcription Factors - Abstract
Non-mesenchymal pancreatic cells are a potential source for cell replacement. Their transdifferentiation can be achieved by triggering epigenetic remodeling through e. g. post-translational modification of histones. Valproic acid, a branched-chain saturated fatty acid with histone deacetylase inhibitor activity, was linked to the expression of key transcription factors of pancreatic lineage in epithelial cells and insulin transcription. However, the potential of valproic acid to cause cellular reprogramming is not fully understood. To shed further light on it we employed next-generation RNA sequencing, real-time PCR, and protein analyses by ELISA and western blot, to assess the impact of valproic acid on transcriptome and function of Panc-1-cells. Our results indicate that valproic acid has a significant impact on the cell cycle, cell adhesion, histone H3 acetylation, and metabolic pathways as well as the initiation of epithelial-mesenchymal transition through acetylation of histone H3 resulting in α-cell-like characteristics. We conclude that human epithelial pancreatic cells can be transdifferentiated into cells with endocrine properties through epigenetic regulation by valproic acid favoring an α-cell-like phenotype.
- Published
- 2022
236. Plasticity of ocular surface epithelia: Using a murine model of limbal stem cell deficiency to delineate metaplasia and transdifferentiation
- Author
-
Mijeong Park, Richard Zhang, Elvis Pandzic, Mingxia Sun, Vivien J. Coulson-Thomas, and Nick Di Girolamo
- Subjects
Metaplasia ,Stem Cells ,Epithelium, Corneal ,Cell Biology ,Limbus Corneae ,Biochemistry ,Corneal Diseases ,Mice ,Disease Models, Animal ,Cell Transdifferentiation ,Genetics ,Humans ,Animals ,Developmental Biology - Abstract
Maintaining corneal health and transparency are necessary pre-requisites for exquisite vision, a function ascribed to stem cells (SCs) nestled within the limbus. Perturbations to this site or depletion of its SCs results in limbal SC deficiency. While characterizing a murine model of this disease, we discovered unusual transformation phenomena on the corneal surface including goblet cell metaplasia (GCM), conjunctival transdifferentiation, and squamous metaplasia (SQM). GCM arose from K8
- Published
- 2022
237. EPO synthesis induced by HIF-PHD inhibition is dependent on myofibroblast transdifferentiation and colocalizes with non-injured nephron segments in murine kidney fibrosis
- Author
-
Hanako Kobayashi, Olena Davidoff, Shiuli Pujari‐Palmer, Malin Drevin, and Volker H. Haase
- Subjects
Mice, Knockout ,Mice ,Physiology ,Cell Transdifferentiation ,Animals ,Nephrons ,Renal Insufficiency, Chronic ,Kidney ,Myofibroblasts ,Erythropoietin ,Fibrosis ,Prolyl Hydroxylases ,Hypoxia-Inducible Factor-Proline Dioxygenases - Abstract
Erythropoietin (EPO) is regulated by hypoxia-inducible factor (HIF)-2. In the kidney, it is produced by cortico-medullary perivascular interstitial cells, which transdifferentiate into collagen-producing myofibroblasts in response to injury. Inhibitors of prolyl hydroxylase domain (PHD) dioxygenases (HIF-PHIs) activate HIF-2 and stimulate kidney and liver EPO synthesis in patients with anemia of chronic kidney disease (CKD). We examined whether HIF-PHIs can reactivate EPO synthesis in interstitial cells that have undergone myofibroblast transdifferentiation in established kidney fibrosis.We investigated Epo transcription in myofibroblasts and characterized the histological distribution of kidney Epo transcripts by RNA in situ hybridization combined with immunofluorescence in mice with adenine nephropathy (AN) treated with HIF-PHI molidustat. Lectin absorption chromatography was used to assess liver-derived EPO. In addition, we examined kidney Epo transcription in Phd2 knockout mice with obstructive nephropathy.In AN, molidustat-induced Epo transcripts were not found in areas of fibrosis and did not colocalize with interstitial cells that expressed α-smooth muscle actin, a marker of myofibroblast transdifferentiation. Epo transcription was associated with megalin-expressing, kidney injury molecule 1-negative nephron segments and contingent on residual renal function. Liver-derived EPO did not contribute to serum EPO in molidustat-treated mice. Epo transcription was not associated with myofibroblasts in Phd2 knockout mice with obstructive nephropathy.Our studies suggest that HIF-PHIs do not reactivate Epo transcription in interstitial myofibroblasts and that their efficacy in inducing kidney EPO in CKD is dependent on the degree of myofibroblast formation, the preservation of renal parenchyma and the level of residual renal function.
- Published
- 2022
238. SARA suppresses myofibroblast precursor transdifferentiation in fibrogenesis in a mouse model of scleroderma
- Author
-
Katia Corano Scheri, Xiaoyan Liang, Vidhi Dalal, I. Caroline Le Poole, John Varga, and Tomoko Hayashida
- Subjects
Mice ,Bleomycin ,Disease Models, Animal ,Scleroderma, Systemic ,Cell Transdifferentiation ,Animals ,General Medicine ,Myofibroblasts ,Fibrosis - Abstract
We previously reported that Smad anchor for receptor activation (SARA) plays a critical role in maintaining epithelial cell phenotype. Here, we show that SARA suppressed myofibroblast precursor transdifferentiation in a mouse model of scleroderma. Mice overexpressing SARA specifically in PDGFR-β+ pericytes and pan-leukocytes (SARATg) developed significantly less skin fibrosis in response to bleomycin injection compared with wild-type littermates (SARAWT). Single-cell RNA-Seq analysis of skin PDGFR-β+ cells implicated pericyte subsets assuming myofibroblast characteristics under fibrotic stimuli, and SARA overexpression blocked the transition. In addition, a cluster that expresses molecules associated with Th2 cells and macrophage activation was enriched in SARAWT mice, but not in SARATg mice, after bleomycin treatment. Th2-specific Il-31 expression was increased in skin of the bleomycin-treated SARAWT mice and patients with scleroderma (or systemic sclerosis, SSc). Receptor-ligand analyses indicated that lymphocytes mediated pericyte transdifferentiation in SARAWT mice, while with SARA overexpression the myofibroblast activity of pericytes was suppressed. Together, these data suggest a potentially novel crosstalk between myofibroblast precursors and immune cells in the pathogenesis of SSc, in which SARA plays a critical role.
- Published
- 2022
239. IN BRAIN POST-ISCHEMIC PLASTICITY, Na
- Author
-
Antonella, Casamassa, Ornella, Cuomo, Anna, Pannaccione, Pasquale, Cepparulo, Giusy, Laudati, Valeria, Valsecchi, Lucio, Annunziato, and Giuseppe, Pignataro
- Subjects
Neurons ,Mice ,Ischemia ,Astrocytes ,Cell Transdifferentiation ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Microglia ,Sodium-Calcium Exchanger ,Brain Ischemia - Abstract
The intricate glia interaction occurring after stroke strongly depend on the maintenance of intraglial ionic homeostasis. Among the several ionic channels and transporters, the plasmamembrane Na
- Published
- 2022
240. Monoamine oxidase A drives neuroendocrine differentiation in prostate cancer
- Author
-
Xue Shui, Xuehua Ren, Rong Xu, Qinghua Xie, Yaohua Hu, Jing Qin, Han Meng, Caiqin Zhang, Jumei Zhao, and Changhong Shi
- Subjects
Male ,Cell Line, Tumor ,Cell Transdifferentiation ,Biophysics ,Humans ,Prostatic Neoplasms ,Cell Biology ,Molecular Biology ,Biochemistry ,Monoamine Oxidase ,Signal Transduction - Abstract
Neuroendocrine transdifferentiation (NED) of prostate cancer (PCa) is the main cause of failure of androgen receptor inhibitor treatment. However, the molecular mechanisms underlying the development of NEPC, especially treatment-induced NEPC, remain unclear. Emerging evidence indicates that elevated monoamine oxidase A (MAOA) contribute to the proliferation, cell stemness, and bone metastasis in PCa. Here, we generated an enzalutamide-induced NED cell model to assess the role of MAOA during NED. Overall, MAOA expression was significantly increased upon Enz long-term exposure and was required for neuroendocrine marker expression. In particular, Enz was found to induce NED via the MAOA/mTOR/HIF-1α signaling axis. Further analyses revealed that the MAOA inhibitor clorgyline(CLG) may bring multiple benefits to CRPC patients, including better therapeutic effect and delays NED. These findings suggest that MAOA may be an important target for the development of anti-NED therapies, thereby providing a novel strategy for the combined application of CLG and AR inhibitors in the clinic.
- Published
- 2022
241. Loss of SUMO-specific protease 2 causes isolated glucocorticoid deficiency by blocking adrenal cortex zonal transdifferentiation in mice
- Author
-
Damien Dufour, Typhanie Dumontet, Isabelle Sahut-Barnola, Aude Carusi, Méline Onzon, Eric Pussard, James Jr Wilmouth, Julie Olabe, Cécily Lucas, Adrien Levasseur, Christelle Damon-Soubeyrand, Jean-Christophe Pointud, Florence Roucher-Boulez, Igor Tauveron, Guillaume Bossis, Edward T. Yeh, David T. Breault, Pierre Val, Anne-Marie Lefrançois-Martinez, and Antoine Martinez
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Cysteine Endopeptidases ,Adrenocorticotropic Hormone ,Adrenal Cortex Hormones ,Cell Transdifferentiation ,Adrenal Cortex ,Animals ,Glucocorticoids ,Wnt Signaling Pathway ,beta Catenin - Abstract
SUMOylation is a dynamic posttranslational modification, that provides fine-tuning of protein function involved in the cellular response to stress, differentiation, and tissue development. In the adrenal cortex, an emblematic endocrine organ that mediates adaptation to physiological demands, the SUMOylation gradient is inversely correlated with the gradient of cellular differentiation raising important questions about its role in functional zonation and the response to stress. Considering that SUMO-specific protease 2 (SENP2), a deSUMOylating enzyme, is upregulated by Adrenocorticotropic Hormone (ACTH)/cAMP-dependent Protein Kinase (PKA) signalling within the zonafasciculata, we generated mice with adrenal-specific Senp2 loss to address these questions. Disruption of SENP2 activity in steroidogenic cells leads to specific hypoplasia of the zona fasciculata, a blunted reponse to ACTH and isolated glucocorticoid deficiency. Mechanistically, overSUMOylation resulting from SENP2 loss shifts the balance between ACTH/PKA and WNT/β-catenin signalling leading to repression of PKA activity and ectopic activation of β-catenin. At the cellular level, this blocks transdifferentiation of β-catenin-positive zona glomerulosa cells into fasciculata cells and sensitises them to premature apoptosis. Our findings indicate that the SUMO pathway is critical for adrenal homeostasis and stress responsiveness.
- Published
- 2022
242. Aorta in Pathologies May Function as an Immune Organ by Upregulating Secretomes for Immune and Vascular Cell Activation, Differentiation and Trans-Differentiation—Early Secretomes may Serve as Drivers for Trained Immunity
- Author
-
Yifan, Lu, Yu, Sun, Keman, Xu, Fatma, Saaoud, Ying, Shao, Charles, Drummer, Sheng, Wu, Wenhui, Hu, Jun, Yu, Satya P, Kunapuli, John R, Bethea, Roberto I, Vazquez-Padron, Jianxin, Sun, Xiaohua, Jiang, Hong, Wang, and Xiaofeng, Yang
- Subjects
NF-E2-Related Factor 2 ,Angiotensin II ,Caspase 1 ,Immunology ,COVID-19 ,Endothelial Cells ,Cell Differentiation ,stomatognathic system ,Cell Transdifferentiation ,Middle East Respiratory Syndrome Coronavirus ,cardiovascular system ,Cytokines ,Humans ,Immunology and Allergy ,Renal Insufficiency, Chronic ,Aorta ,Secretome - Abstract
To determine whether aorta becomes immune organ in pathologies, we performed transcriptomic analyses of six types of secretomic genes (SGs) in aorta and vascular cells and made the following findings: 1) 53.7% out of 21,306 human protein genes are classified into six secretomes, namely, canonical, caspase 1, caspase 4, exosome, Weibel–Palade body, and autophagy; 2) Atherosclerosis (AS), chronic kidney disease (CKD) and abdominal aortic aneurysm (AAA) modulate six secretomes in aortas; and Middle East Respiratory Syndrome Coronavirus (MERS-CoV, COVID-19 homologous) infected endothelial cells (ECs) and angiotensin-II (Ang-II) treated vascular smooth muscle cells (VSMCs) modulate six secretomes; 3) AS aortas upregulate T and B cell immune SGs; CKD aortas upregulate SGs for cardiac hypertrophy, and hepatic fibrosis; and AAA aorta upregulate SGs for neuromuscular signaling and protein catabolism; 4) Ang-II induced AAA, canonical, caspase 4, and exosome SGs have two expression peaks of high (day 7)-low (day 14)-high (day 28) patterns; 5) Elastase induced AAA aortas have more inflammatory/immune pathways than that of Ang-II induced AAA aortas; 6) Most disease-upregulated cytokines in aorta may be secreted via canonical and exosome secretomes; 7) Canonical and caspase 1 SGs play roles at early MERS-CoV infected ECs whereas caspase 4 and exosome SGs play roles in late/chronic phases; and the early upregulated canonical and caspase 1 SGs may function as drivers for trained immunity (innate immune memory); 8) Venous ECs from arteriovenous fistula (AVF) upregulate SGs in five secretomes; and 9) Increased some of 101 trained immunity genes and decreased trained tolerance regulator IRG1 participate in upregulations of SGs in atherosclerotic, Ang-II induced AAA and CKD aortas, and MERS-CoV infected ECs, but less in SGs upregulated in AVF ECs. IL-1 family cytokines, HIF1α, SET7 and mTOR, ROS regulators NRF2 and NOX2 partially regulate trained immunity genes; and NRF2 plays roles in downregulating SGs more than that of NOX2 in upregulating SGs. These results provide novel insights on the roles of aorta as immune organ in upregulating secretomes and driving immune and vascular cell differentiations in COVID-19, cardiovascular diseases, inflammations, transplantations, autoimmune diseases and cancers.
- Published
- 2022
- Full Text
- View/download PDF
243. 共培养方法诱导3种间充质干细胞向神经细胞分化的比较.
- Author
-
徐丽丽, 工洪元, 李学达, 刘兵, 郑方芳, and 杨乃龙
- Abstract
BACKGROUND: Scholars have been trying to create a microenvironment similar to the human body, which can induce the directional differentiation of mesenchymal stem cells from human bone marrow, placenta and umbilical cord blood. OBJECTIVE: To compare the neuronal differentiation of human bone marrow mesenchymal stem cells, human placental mesenchymal stem cells and human umbilical cord blood mesenchymal stem cells induced by co-culture with nerve cells. METHODS: Human bone marrow mesenchymal stem cells, human placental mesenchymal stem cells and human umbilical cord blood mesenchymal stem cells cultured in vitro were co-cultured with nerve cells using the Transwell system. The morphological changes of three kinds of cells in the co-culture system were detected. After co-culture for 4-5 days, immunofluorescence staining was used to measure the expression of neuron-specific enolase in cells. Mesenchymal stem cells only cultured in low glucose DMEM medium were used as controls. RESULTS AND CONCLUSION: These three kinds of mesenchymal stem cells were extended, and interconnected processes were detective. The positive expression of neuron-specific enolase was highest in the human umbilical cord blood mesenchymal stem cells followed by human placental mesenchymal stem cells and human bone marrow mesenchymal stem cells in order. In the control group, none of the three kinds of mesenchymal stem cells have neuronal morphology, and the expression of neuron specific enolase was negative for the immunofluorescence staining. To conclude, microenvironment provided by nerve cells can induce these three kinds of mesenchymal stem cells to differentiate into neurons. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
244. Myokardiales Tissue-Engineering.
- Author
-
Martin, U. and Haverich, A.
- Abstract
Copyright of Zeitschrift für Herz-, Thorax- und Gefaesschirurgie is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2017
- Full Text
- View/download PDF
245. The orphan nuclear receptor COUP-TFII coordinates hypoxia-independent proangiogenic responses in hepatic stellate cells.
- Author
-
Tempesti, Sara, Marroncini, Giada, Ceni, Elisabetta, Mello, Tommaso, Polvani, Simone, Tarocchi, Mirko, Milani, Stefano, Galli, Andrea, Vasseur-Cognet, Mireille, Cavalieri, Duccio, Beltrame, Luca, and Pinzani, Massimo
- Subjects
- *
LIVER cells , *NUCLEAR receptors (Biochemistry) , *OVALBUMINS , *MYOFIBROBLASTS , *NEOVASCULARIZATION - Abstract
Background & Aims Hepatic stellate cell (HSC) transdifferentiation into collagen-producing myofibroblasts is a key event in hepatic fibrogenesis, but the transcriptional network that controls the acquisition of the activated phenotype is still poorly understood. In this study, we explored whether the nuclear receptor chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) is involved in HSC activation and in the multifunctional role of these cells during the response to liver injury. Methods COUP-TFII expression was evaluated in normal and cirrhotic livers by immunohistochemistry and Western blot. The role of COUP-TFII in HSC was assessed by gain and loss of function transfection experiments and by generation of mice with COUP-TFII deletion in HSC. Molecular changes were determined by gene expression microarray and RT-qPCR. Results We showed that COUP-TFII is highly expressed in human fibrotic liver and in mouse models of hepatic injury. COUP-TFII expression rapidly increased upon HSC activation and it was associated with the regulation of genes involved in cell motility, proliferation and angiogenesis. Inactivation of COUP-TFII impairs proliferation and invasiveness in activated HSC and COUP-TFII deletion in mice abrogate HSC activation and angiogenesis. Finally, co-culture experiments with HSC and liver sinusoidal endothelial cells (SEC) showed that COUP-TFII expression in HSC influenced SEC migration and tubulogenesis via a hypoxia-independent and nuclear factor kappaB-dependent mechanism. Conclusion This study elucidates a novel transcriptional pathway in HSC that is involved in the acquisition of the proangiogenic phenotype and regulates the paracrine signals between HSC and SEC during hepatic wound healing. Lay summary In this study, we identified an important regulator of HSC pathobiology. We showed that the orphan receptor COUP-TFII is an important player in hepatic neoangiogenesis. COUP-TFII expression in HSC controls the crosstalk between HSC and endothelial cells coordinating vascular remodelling during liver injury. Transcript profiling ArrayExpress accession E-MTAB-1795. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
246. The Conservation of the Germline Multipotency Program, from Sponges to Vertebrates: A Stepping Stone to Understanding the Somatic and Germline Origins.
- Author
-
Fierro-Constaín, Laura, Schenkelaars, Quentin, Gazave, Eve, Haguenauer, Anne, Rocher, Caroline, Ereskovsky, Alexander, Borchiellini, Carole, and Renard, Emmanuelle
- Subjects
- *
GERM cells , *SOMATIC embryogenesis , *SPONGES (Invertebrates) , *VERTEBRATES , *CTENOPHORA , *METAZOA - Abstract
The germline definition in metazoans was first based on few bilaterian models. As a result, gene function interpretations were often based on phenotypes observed in those models and led to the definition of a set of genes, considered as specific of the germline, named the "germline core". However, some of these genes were shown to also be involved in somatic stem cells, thus leading to the notion of germline multipotency program (GMP). Because Porifera and Ctenophora are currently the best candidates to be the sister-group to all other animals, the comparative analysis of gene contents and functions between these phyla, Cnidaria and Bilateria is expected to provide clues on early animal evolution and on the links between somatic and germ lineages. Our present bioinformatic analyses at the metazoan scale show that a set of 18 GMP genes was already present in the last common ancestor of metazoans and indicate more precisely the evolution of some of them in the animal lineage. The expression patterns and levels of 11 of these genes in the homoscleromorph sponge Oscarella lobularis show that they are expressed throughout their life cycle, in pluri/multipotent progenitors, during gametogenesis, embryogenesis and during wound healing. This new study in a nonbilaterian species reinforces the hypothesis of an ancestral multipotency program. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
247. Roles of Chondrocytes in Endochondral Bone Formation and Fracture Repair.
- Author
-
Hinton, R. J., Jing, Y., Jing, J., and Feng, J. Q.
- Subjects
CARTILAGE cells ,ENDOCHONDRAL ossification ,BONE growth ,OSTEOBLASTS ,TEMPOROMANDIBULAR joint ,BONE cells ,THERAPEUTICS ,ANIMALS ,BONE fractures ,FRACTURE healing ,PHYSIOLOGY - Abstract
The formation of the mandibular condylar cartilage (MCC) and its subchondral bone is an important but understudied topic in dental research. The current concept regarding endochondral bone formation postulates that most hypertrophic chondrocytes undergo programmed cell death prior to bone formation. Under this paradigm, the MCC and its underlying bone are thought to result from 2 closely linked but separate processes: chondrogenesis and osteogenesis. However, recent investigations using cell lineage tracing techniques have demonstrated that many, perhaps the majority, of bone cells are derived via direct transformation from chondrocytes. In this review, the authors will briefly discuss the history of this idea and describe recent studies that clearly demonstrate that the direct transformation of chondrocytes into bone cells is common in both long bone and mandibular condyle development and during bone fracture repair. The authors will also provide new evidence of a distinct difference in ossification orientation in the condylar ramus (1 ossification center) versus long bone ossification formation (2 ossification centers). Based on our recent findings and those of other laboratories, we propose a new model that contrasts the mode of bone formation in much of the mandibular ramus (chondrocyte-derived) with intramembranous bone formation of the mandibular body (non-chondrocyte-derived). [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
248. Sox10 escalates vascular inflammation by mediating vascular smooth muscle cell transdifferentiation and pyroptosis in neointimal hyperplasia.
- Author
-
Xu X, Zhang DD, Kong P, Gao YK, Huang XF, Song Y, Zhang WD, Guo RJ, Li CL, Chen BW, Sun Y, Zhao YB, Jia FY, Wang X, Zhang F, and Han M
- Subjects
- Mice, Animals, Hyperplasia pathology, Cell Proliferation physiology, Pyroptosis, Phosphatidylinositol 3-Kinases metabolism, Cell Transdifferentiation, Neointima metabolism, Neointima pathology, Mice, Knockout, Inflammation pathology, Myocytes, Smooth Muscle metabolism, Cells, Cultured, Cell Movement, SOXE Transcription Factors genetics, SOXE Transcription Factors metabolism, Muscle, Smooth, Vascular metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Vascular smooth muscle cells (VSMCs) can transdifferentiate into macrophage-like cells in the context of sustained inflammatory injury, which drives vascular hyperplasia and atherosclerotic complications. Using single-cell RNA sequencing, we identify that macrophage-like VSMCs are the key cell population in mouse neointimal hyperplasia. Sex-determining region Y (SRY)-related HMG-box gene 10 (Sox10) upregulation is associated with macrophage-like VSMC accumulation and pyroptosis in vitro and in the neointimal hyperplasia of mice. Tumor necrosis factor α (TNF-α)-induced Sox10 lactylation in a phosphorylation-dependent manner by PI3K/AKT signaling drives transcriptional programs of VSMC transdifferentiation, contributing to pyroptosis. The regulator of G protein signaling 5 (RGS5) interacts with AKT and blocks PI3K/AKT signaling and Sox10 phosphorylation at S24. Sox10 silencing mitigates vascular inflammation and forestalls neointimal hyperplasia in RGS5 knockout mice. Collectively, this study shows that Sox10 is a regulator of vascular inflammation and a potential control point in inflammation-related vascular disease., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
249. Electric field promotes dermal fibroblast transdifferentiation through activation of RhoA/ROCK1 pathway.
- Author
-
Wang W, Huang W, Liu J, Zhang Z, Ji R, Wu C, Zhang J, and Jiang X
- Subjects
- Signal Transduction, Wound Healing, Cell Transdifferentiation, Fibroblasts metabolism
- Abstract
With the increased incidence of age-related and lifestyle-related diseases, chronic wounds are sweeping the world, where recent studies reveal that dysfunction of fibroblast plays an indispensable role. Endogenous electric field (EF) generated by skin wound disrupting an epithelial layer has been used as an alternative clinical treatment in chronic wound by modulating cellular behaviours, including fibroblasts transdifferentiation. Although many molecules and signaling pathways have been reported associated with fibroblasts transdifferentiation, studies investigating how the electric field affects the cellular pathways have been limited. For this purpose, a model of electric field treatment in vitro was established, where cells were randomly divided into control and electrified groups. The changes of protein expression and distribution were detected under different conditions, along with Zeiss imaging system observing the response of cells. Results showed that fibroblast transdifferentiation was accompanied by increased expression of a-SMA and extracellular matrix (COL-1 and COL-3) under the EF. Simultaneously, fibroblast transdifferentiation was also consistent with changes of cell arrangement and enhanced motility. Furthermore, we found that electric field activated RhoA signaling pathways activity. Y-27632, a RhoA inhibitor, which was used to treat fibroblasts, resulted in reduced transdifferentiation. The connection between electric field and RhoA signaling pathways is likely to be significant in modulating fibroblast transdifferentiation in acute injury and tissue remodeling, which provides an innovative idea for the molecular mechanism of EF in promoting chronic wound healing., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2023
- Full Text
- View/download PDF
250. Transdifferentiation of B-lymphoblastic leukemia to histiocytic sarcoma after immunotherapy.
- Author
-
Fenu EM, Margolskee E, and Pillai V
- Subjects
- Humans, Cell Transdifferentiation, Immunotherapy, Histiocytic Sarcoma therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma, Lymphoma, B-Cell
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.