Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30.202
Filtrar
1.
Cells ; 13(9)2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38727314

RESUMO

During the secretory phase of the menstrual cycle, endometrial fibroblast cells begin to change into large epithelial-like cells called decidual cells in a process called decidualization. This differentiation continues more broadly in the endometrium and forms the decidual tissue during early pregnancy. The cells undergoing decidualization as well as the resulting decidual cells, support successful implantation and placentation during early pregnancy. This study was carried out to identify new potentially important long non-coding RNA (lncRNA) genes that may play a role in human endometrial stromal fibroblast cells (hESF) undergoing decidualization in vitro, and several were found. The expression of nine was further characterized. One of these, AC027288.3, showed a dramatic increase in the expression of hESF cells undergoing decidualization. When AC027288.3 expression was targeted, the ability of the cells to undergo decidualization as determined by the expression of decidualization marker protein-coding genes was significantly altered. The most affected markers of decidualization whose expression was significantly reduced were FOXO1, FZD4, and INHBA. Therefore, AC027288.3 may be a major upstream regulator of the WNT-FOXO1 pathway and activin-SMAD3 pathways previously shown as critical for hESF decidualization. Finally, we explored possible regulators of AC027288.3 expression during human ESF decidualization. Expression was regulated by cAMP and progesterone. Our results suggest that AC027288.3 plays a role in hESF decidualization and identifies several other lncRNA genes that may also play a role.


Assuntos
Decídua , Endométrio , Fibroblastos , RNA Longo não Codificante , Células Estromais , Humanos , Feminino , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fibroblastos/metabolismo , Fibroblastos/citologia , Decídua/metabolismo , Decídua/citologia , Endométrio/citologia , Endométrio/metabolismo , Células Estromais/metabolismo , Células Estromais/citologia , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Gravidez , Adulto , Diferenciação Celular/genética
2.
Stem Cell Res Ther ; 15(1): 129, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38693588

RESUMO

BACKGROUND: Human bone marrow-derived stem cells (hBMDSCs) are well characterized mediators of tissue repair and regeneration. An increasing body of evidence indicates that these cells exert their therapeutic effects largely through their paracrine actions rather than clonal expansion and differentiation. Here we studied the role of microRNAs (miRNAs) present in extracellular vesicles (EVs) from hBMDSCs in tissue regeneration and cell differentiation targeting endometrial stromal fibroblasts (eSF). METHODS: Extracellular vesicles (EVs) are isolated from hBMDSCs, characterized by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) techniques. Extracted total RNA from EVs was subjected to RNA seq analysis. Transfection and decidualization studies were carried out in endometrial stromal fibroblasts (eSF). Gene expression was analyzed by qRTPCR. Unpaired t-test with Welch's correction was used for data analysis between two groups. RESULTS: We identified several microRNAs (miRNAs) that were highly expressed, including miR-21-5p, miR-100-5p, miR-143-3p and let7. MiR-21 is associated with several signaling pathways involved in tissue regeneration, quiescence, cellular senescence, and fibrosis. Both miR-100-5p and miR-143-3p promoted cell proliferation. MiR-100-5p specifically promoted regenerative processes by upregulating TGF-ß3, VEGFA, MMP7, and HGF. MiR-100-5p blocked differentiation or decidualization as evidenced by morphologic changes and downregulation of decidualization mediators including HOXA10, IGFBP1, PRL, PR-B, and PR. CONCLUSION: EVs delivered to tissues by hBMDSCs contain specific miRNAs that prevent terminal differentiation and drive repair and regeneration. Delivery of microRNAs is a novel treatment paradigm with the potential to replace BMDSCs in cell-free regenerative therapies.


Assuntos
Diferenciação Celular , Proliferação de Células , Endométrio , Exossomos , Fibroblastos , Células-Tronco Mesenquimais , MicroRNAs , Humanos , MicroRNAs/metabolismo , MicroRNAs/genética , Feminino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Exossomos/metabolismo , Endométrio/metabolismo , Endométrio/citologia , Fibroblastos/metabolismo , Fibroblastos/citologia , Regeneração/genética , Células da Medula Óssea/metabolismo , Células da Medula Óssea/citologia
3.
J Nanobiotechnology ; 22(1): 220, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38698449

RESUMO

BACKGROUND: Bone marrow mesenchymal stem cells (BMSCs) can differentiate into Schwann cells (SCs) during peripheral nerve injury; in our previous research, we showed that SC-derived exosomes (SC-exos) played a direct induction role while fibroblast-derived exosomes (Fb-exos) had no obvious induction role. The induction role of neural stem cell (NSC)-derived exosomes (NSC-exos) has also been widely confirmed. However, no studies have compared the induction effects of these three types of cells at the same time. Therefore, by investigating the effect of these three cell-derived exosomes upon the induction of BMSCs to differentiate into SCs, this study explored the role of different exosomes in promoting the differentiation of stem cells into SCs cells, and conducted a comparison between the two groups by RNA sequencing to further narrow the range of target genes and related gene pathways in order to study their related mechanisms. MATERIALS AND METHODS: We extracted exosomes from SCs, fibroblasts (Fb) and neural stem cells (NSC) and then investigated the ability of these exosomes to induce differentiation into BMSCs under different culture conditions. The expression levels of key proteins and gene markers were detected in induced cells by fluorescence immunoassays, western blotting and polymerase chain reaction (PCR); then, we statistically compared the relative induction effects under different conditions. Finally, we analyzed the three types of exosomes by RNA-seq to predict target genes and related gene pathways. RESULTS: BMSCs were cultured by three media: conventional (no induction), pre-induction or pre-induction + original induction medium (ODM) with exosomes of the same cell origin under different culture conditions. When adding the three different types of exosomes separately, the overall induction of BMSCs to differentiate into SCs was significantly increased (P < 0.05). The induction ability was ranked as follows: pre-induction + ODM + exosome group > pre-induction + exosome group > non-induction + exosome group. Using exosomes from different cell sources under the same culture conditions, we observed the following trends under the three culture conditions: RSC96-exos group ≥ NSC-exos group > Fb-exos group. The overall ability to induce BMSCs into SCs was significantly greater in the RSC96-exos group and the NSC-exos group. Although there was no significant difference in induction efficiency when comparing these two groups, the overall induction ability of the RSC96-exos group was slightly higher than that of the NSC-exos group. By combining the differentiation induction results with the RNA-seq data, the three types of exosomes were divided into three comparative groups: RSC vs. NSC, RSC vs. Fb and NSC vs. Fb. We identified 203 differentially expressed mRNA target genes in these three groups. Two differentially expressed genes were upregulated simultaneously, namely riboflavin kinase (RFK, ENSRNOG00000022273) and ribosomal RNA processing 36 (Rrp36, ENSRNOG00000017836). We did not identify any co-upregulated target genes for the miRNAs, but did identify one target gene of the lncRNAs, namely ENSRNOG00000065005. Analysis identified 90 GO terms related to nerves and axons in the mRNAs; in addition, KEGG enrichment and GASA analysis identified 13 common differential expression pathways in the three groups. CONCLUSIONS: Our analysis found that pre-induction + ODM + RSC96/NSC-exos culture conditions were most conducive with regards to induction and differentiation. RSC96-exos and NSC-exos exhibited significantly greater differentiation efficiency of BMSCs into SCs. Although there was no statistical difference, the data indicated a trend for RSC96-exos to be advantageous We identified 203 differentially expressed mRNAs between the three groups and two differentially expressed target mRNAs were upregulated, namely riboflavin kinase (RFK, ENSRNOG00000022273) and ribosomal RNA processing 36 (Rrp36, ENSRNOG00000017836). 90 GO terms were related to nerves and axons. Finally, we identified 13 common differentially expressed pathways across our three types of exosomes. It is hoped that the efficiency of BMSCs induction differentiation into SCs can be improved, bringing hope to patients and more options for clinical treatment.


Assuntos
Diferenciação Celular , Exossomos , Células-Tronco Mesenquimais , Células de Schwann , Exossomos/metabolismo , Células de Schwann/citologia , Células de Schwann/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Ratos , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Ratos Sprague-Dawley , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo
4.
Curr Protoc ; 4(5): e1012, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38712688

RESUMO

Although protocols to generate authentic transgene-free mouse and human induced pluripotent stem cells (iPSCs) are now well established, standard methods for reprogramming porcine somatic cells still suffer from low efficiency and transgene retention. The Basic Protocol describes reprogramming procedures to establish transgene-free porcine iPSCs (PiPSCs) from porcine fibroblasts. This method uses episomal plasmids encoding POU5F1, SOX2, NANOG, KLF4, SV40LT, c-MYC, LIN28A, and microRNA-302/367, combined with an optimized medium, to establish PiPSC lines. Support protocols describe the establishment and characterization of clonal PiPSC lines, as well as the preparation of feeder cells and EBNA1 mRNA. This optimized, step-by-step approach tailored to this species enables the efficient derivation of PiPSCs in ∼4 weeks. The establishment of transgene-free PiPSCs provides a new and valuable model for studies of larger mammalian species' development, disease, and regenerative biology. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Reprogramming of porcine fibroblasts with episomal plasmids Support Protocol 1: Preparation of mouse embryonic fibroblasts for feeder layer Support Protocol 2: Preparation of in vitro-transcribed EBNA1 mRNA Support Protocol 3: Establishment of clonal porcine induced pluripotent stem cell (PiPSC) lines Support Protocol 4: PiPSC characterization: Genomic DNA PCR and RT-PCR Support Protocol 5: PiPSC characterization: Immunostaining.


Assuntos
Células-Tronco Pluripotentes Induzidas , Fator 4 Semelhante a Kruppel , Transgenes , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Suínos , Camundongos , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Cultura de Células/métodos , Reprogramação Celular/genética
5.
Sci Rep ; 14(1): 10295, 2024 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-38704415

RESUMO

Lysine crotonylation (Kcr) is a recently discovered histone acylation modification that is closely associated with gene expression, cell proliferation, and the maintenance of stem cell pluripotency and indicates the transcriptional activity of genes and the regulation of various biological processes. During cell culture, the introduction of exogenous croconic acid disodium salt (Nacr) has been shown to modulate intracellular Kcr levels. Although research on Kcr has increased, its role in cell growth and proliferation and its potential regulatory mechanisms remain unclear compared to those of histone methylation and acetylation. Our investigation demonstrated that the addition of 5 mM Nacr to cultured bovine fibroblasts increased the expression of genes associated with Kcr modification, ultimately promoting cell growth and stimulating cell proliferation. Somatic cell nuclear transfer of donor cells cultured in 5 mM Nacr resulted in 38.1% blastocyst development, which was significantly greater than that in the control group (25.2%). This research is important for elucidating the crotonylation modification mechanism in fibroblast proliferation to promote the efficacy of somatic cell nuclear transfer.


Assuntos
Proliferação de Células , Fibroblastos , Histonas , Técnicas de Transferência Nuclear , Animais , Bovinos , Fibroblastos/metabolismo , Fibroblastos/citologia , Proliferação de Células/efeitos dos fármacos , Histonas/metabolismo , Desenvolvimento Embrionário , Blastocisto/metabolismo , Blastocisto/citologia , Lisina/metabolismo , Crotonatos/metabolismo , Células Cultivadas , Processamento de Proteína Pós-Traducional , Feminino
6.
Sci Rep ; 14(1): 10365, 2024 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710778

RESUMO

Cardiac fibroblasts (CFs) are essential for preserving myocardial integrity and function. They can detect variations in cardiac tissue stiffness using various cellular mechanosensors, including the Ca2+ permeable mechanosensitive channel Piezo1. Nevertheless, how CFs adapt the mechanosensitive response to stiffness changes remains unclear. In this work we adopted a multimodal approach, combining the local mechanical stimulation (from 10 pN to 350 nN) with variations of culture substrate stiffness. We found that primary rat CFs cultured on stiff (GPa) substrates showed a broad Piezo1 distribution in the cell with particular accumulation at the mitochondria membrane. CFs displayed a force-dependent behavior in both calcium uptake and channel activation probability, showing a threshold at 300 nN, which involves both cytosolic and mitochondrial Ca2+ mobilization. This trend decreases as the myofibroblast phenotype within the cell population increases, following a possible Piezo1 accumulation at focal adhesion sites. In contrast, the inhibition of fibroblasts to myofibroblasts transition with soft substrates (kPa) considerably reduces both mechanically- and chemically-induced Piezo1 activation and expression. Our findings shed light on how Piezo1 function and expression are regulated by the substrate stiffness and highlight its involvement in the environment-mediated modulation of CFs mechanosensitivity.


Assuntos
Fibroblastos , Canais Iônicos , Mecanotransdução Celular , Proteínas de Membrana , Animais , Canais Iônicos/metabolismo , Ratos , Fibroblastos/metabolismo , Fibroblastos/citologia , Células Cultivadas , Cálcio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Miocárdio/metabolismo , Miocárdio/citologia , Microambiente Celular
7.
Am J Hum Genet ; 111(5): 841-862, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38593811

RESUMO

RNA sequencing (RNA-seq) has recently been used in translational research settings to facilitate diagnoses of Mendelian disorders. A significant obstacle for clinical laboratories in adopting RNA-seq is the low or absent expression of a significant number of disease-associated genes/transcripts in clinically accessible samples. As this is especially problematic in neurological diseases, we developed a clinical diagnostic approach that enhanced the detection and evaluation of tissue-specific genes/transcripts through fibroblast-to-neuron cell transdifferentiation. The approach is designed specifically to suit clinical implementation, emphasizing simplicity, cost effectiveness, turnaround time, and reproducibility. For clinical validation, we generated induced neurons (iNeurons) from 71 individuals with primary neurological phenotypes recruited to the Undiagnosed Diseases Network. The overall diagnostic yield was 25.4%. Over a quarter of the diagnostic findings benefited from transdifferentiation and could not be achieved by fibroblast RNA-seq alone. This iNeuron transcriptomic approach can be effectively integrated into diagnostic whole-transcriptome evaluation of individuals with genetic disorders.


Assuntos
Transdiferenciação Celular , Fibroblastos , Neurônios , Análise de Sequência de RNA , Humanos , Transdiferenciação Celular/genética , Fibroblastos/metabolismo , Fibroblastos/citologia , Análise de Sequência de RNA/métodos , Neurônios/metabolismo , Neurônios/citologia , Transcriptoma , Reprodutibilidade dos Testes , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/diagnóstico , RNA-Seq/métodos , Feminino , Masculino
8.
J Biomed Mater Res B Appl Biomater ; 112(5): e35408, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38676958

RESUMO

Gelatin methacrylate (GelMA) hydrogels are expected to be ideal skin tissue engineering dressings for a wide range of clinical treatments. Herein, we report the preparation of GelMA or antifreeze GelMA hydrogel sheets with different GelMA concentrations, crosslinking times, and cryoprotectant (CPA) concentrations. The crystallization properties of GelMA or antifreeze GelMA hydrogel sheets were studied by cryomicroscopy and differential scanning calorimetry (DSC). It was found that the growth of ice crystals was slower when GelMA hydrogel concentration was more than 7%. The 10% DMSO-7% GelMA hydrogel sheets crosslinked for 60 min showed no ice crystal formation and growth during cooling and warming. The DSC results showed that the vitrification temperature of the 10% DMSO-7% GelMA hydrogel sheet was -111°C. Furthermore, slow freezing and rapid freezing of fibroblast-laden GelMA or antifreeze GelMA hydrogel sheets, and tissue-engineered skin constructs were studied. The results showed no significant difference in cell survival between slow (88.8% ± 1.51) and rapid (89.2% ± 3.00) freezing of fibroblast-loaded 10% DMSO-7% GelMA hydrogel sheets, and significantly higher than that of 7% GelMA hydrogel sheets (33.4% ± 5.46). The cell viability was higher in tissue-engineered skin constructs after slow freezing (86.34% ± 1.45) than rapid freezing (72.74% ± 1.34). We believe that the combination of antifreeze hydrogels and tissue engineering will facilitate the cryopreservation of tissue engineering constructs.


Assuntos
Criopreservação , Fibroblastos , Gelatina , Hidrogéis , Engenharia Tecidual , Hidrogéis/química , Hidrogéis/farmacologia , Gelatina/química , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Cristalização , Crioprotetores/farmacologia , Crioprotetores/química , Metacrilatos/química , Pele/metabolismo , Camundongos , Proteínas Anticongelantes/química , Proteínas Anticongelantes/farmacologia , Humanos , Sobrevivência Celular/efeitos dos fármacos
9.
Sci Rep ; 14(1): 9444, 2024 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658667

RESUMO

One of the biggest challenges in tissue engineering and regenerative medicine is to ensure oxygen supply of cells in the (temporary) absence of vasculature. With the vision to exploit photosynthetic oxygen production by microalgae, co-cultivated in close vicinity to oxygen-consuming mammalian cells, we are searching for culture conditions that are compatible for both sides. Herein, we investigated the impact of long-term illumination on mammalian cells which is essential to enable photosynthesis by microalgae: four different cell types-primary human fibroblasts, dental pulp stem cells, and osteoblasts as well as the murine beta-cell line INS-1-were continuously exposed to warm white light, red or blue light over seven days. We observed that illumination with red light has no adverse effects on viability, metabolic activity and growth of the cells whereas exposure to white light has deleterious effects that can be attributed to its blue light portion. Quantification of intracellular glutathione did not reveal a clear correlation of this effect with an enhanced production of reactive oxygen species. Finally, our data indicate that the cytotoxic effect of short-wavelength light is predominantly a direct effect of cell illumination; photo-induced changes in the cell culture media play only a minor role.


Assuntos
Fibroblastos , Luz , Espécies Reativas de Oxigênio , Humanos , Animais , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Fibroblastos/citologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Sobrevivência Celular/efeitos da radiação , Polpa Dentária/citologia , Polpa Dentária/efeitos da radiação , Osteoblastos/metabolismo , Osteoblastos/efeitos da radiação , Osteoblastos/citologia , Células Cultivadas , Linhagem Celular , Células-Tronco/metabolismo , Células-Tronco/efeitos da radiação , Células-Tronco/citologia , Glutationa/metabolismo
10.
Biomaterials ; 308: 122560, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38603826

RESUMO

Cells assemble fibronectin, the major extracellular matrix (ECM) protein, into fibrillar matrices, which serve as 3D architectural scaffolds to provide, together with other ECM proteins tissue-specific environments. Although recent approaches enable to bioengineer 3D fibrillar fibronectin matrices in vitro, it remains elusive how fibronectin can be co-assembled with other ECM proteins into complex 3D fibrillar matrices that recapitulate tissue-specific compositions and cellular responses. Here, we introduce the engineering of fibrillar fibronectin-templated 3D matrices that can be complemented with other ECM proteins, including vitronectin, collagen, and laminin to resemble ECM architectures observed in vivo. For the co-assembly of different ECM proteins, we employed their innate fibrillogenic mechanisms including shear forces, pH-dependent electrostatic interactions, or specific binding domains. Through recapitulating various tissue-specific ECM compositions and morphologies, the large scale multi-composite 3D fibrillar ECM matrices can guide fibroblast adhesion, 3D fibroblast tissue formation, or tissue morphogenesis of epithelial cells. In other examples, we customize multi-composite 3D fibrillar matrices to support the growth of signal propagating neuronal networks and of human brain organoids. We envision that these 3D fibrillar ECM matrices can be tailored in scale and composition to modulate tissue-specific responses across various biological length scales and systems, and thus to advance manyfold studies of cell biological systems.


Assuntos
Matriz Extracelular , Fibroblastos , Fibronectinas , Engenharia Tecidual , Fibronectinas/química , Fibronectinas/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Humanos , Engenharia Tecidual/métodos , Fibroblastos/metabolismo , Fibroblastos/citologia , Animais , Alicerces Teciduais/química , Adesão Celular , Camundongos , Organoides/metabolismo , Organoides/citologia
11.
Sci Rep ; 14(1): 9795, 2024 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684844

RESUMO

Cardiac fibrosis contributes to the development of heart failure, and is the response of cardiac fibroblasts (CFs) to pressure or volume overload. Limiting factors in CFs research are the poor availability of human cells and the tendency of CFs to transdifferentiate into myofibroblasts when cultured in vitro. The possibility to generate CFs from induced pluripotent stem cells (iPSC), providing a nearly unlimited cell source, opens new possibilities. However, the behaviour of iPSC-CFs under mechanical stimulation has not been studied yet. Our study aimed to assess the behaviour of iPSC-CFs under mechanical stretch and pro-fibrotic conditions. First, we confirm that iPSC-CFs are comparable to primary CFs at gene, protein and functional level. Furthermore, iPSC-derived CFs adopt a pro-fibrotic response to transforming growth factor beta (TGF-ß). In addition, mechanical stretch inhibits TGF-ß-induced fibroblast activation in iPSC-CFs. Thus, the responsiveness to cytokines and mechanical stimulation of iPSC-CFs demonstrates they possess key characteristics of primary CFs and may be useful for disease modelling.


Assuntos
Fibroblastos , Células-Tronco Pluripotentes Induzidas , Fator de Crescimento Transformador beta , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Humanos , Fibroblastos/metabolismo , Fibroblastos/citologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Estresse Mecânico , Células Cultivadas , Diferenciação Celular , Miocárdio/citologia , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/citologia , Fibrose
12.
J Neurosci Methods ; 406: 110114, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38522633

RESUMO

BACKGROUND: Induced pluripotent stem cells (iPSCs) derived neural stem cells (NSCs) provide a potential for autologous neural transplantation therapy following neurological insults. Thus far, in preclinical studies the donor iPSCs-NSCs are mostly of human or mouse origin with concerns centering around graft rejection when applied to rat brain injury models. For better survival and integration of transplanted cells in the injured brain in rat models, use of rat-iPSC-NSCs and in combination with biomaterials is of advantageous. Herein, we report a detailed method in generating rat iPSCs with improved reprogramming efficiency and differentiation into neurons. NEW METHOD: Rat fibroblasts were reprogrammed into iPSCs with polybrene and EF1α-STEMCCA-LoxP lentivirus vector. Pluripotency characterization, differentiation into neuronal linage cells were assessed with RT-qPCR, Western blotting, immunostaining and patch-clamp methods. Cells were cultured in a custom-designed integrin array system as well as in a hydrogel-based 3D condition. RESULTS: We describe a thorough method for the generation of rat-iPSC-NSCs, and identify integrin αvß8 as a substrate for the optimal growth of rat-iPSC-NSCs. Furthermore, with hydrogel as the supporting biomaterial in the 3-D culture, when combined with integrin αvß8 binding peptide, it forms a conducive environment for optimal growth and differentiation of iPSC-NSCs into mature neurons. COMPARISON WITH EXISTING METHODS: Published studies about rat-iPSC-NSCs are rare. This study provides a detailed protocol for the generation of rat iPSC-NSCs and optimal growth conditions for neuronal differentiation. Our method is useable for studies to assess the utility of rat iPSC-NSCs for neural transplantation in rat brain injury models.


Assuntos
Diferenciação Celular , Fibroblastos , Células-Tronco Pluripotentes Induzidas , Neurônios , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Fibroblastos/fisiologia , Fibroblastos/citologia , Neurônios/citologia , Neurônios/fisiologia , Diferenciação Celular/fisiologia , Ratos , Células Cultivadas , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Técnicas de Cultura de Células/métodos , Ratos Sprague-Dawley
13.
J Virol ; 98(3): e0156323, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38323811

RESUMO

Macrophages are important target cells for diverse viruses and thus represent a valuable system for studying virus biology. Isolation of primary human macrophages is done by culture of dissociated tissues or from differentiated blood monocytes, but these methods are both time consuming and result in low numbers of recovered macrophages. Here, we explore whether macrophages derived from human induced pluripotent stem cells (iPSCs)-which proliferate indefinitely and potentially provide unlimited starting material-could serve as a faithful model system for studying virus biology. Human iPSC-derived monocytes were differentiated into macrophages and then infected with HIV-1, dengue virus, or influenza virus as model human viruses. We show that iPSC-derived macrophages support the replication of these viruses with kinetics and phenotypes similar to human blood monocyte-derived macrophages. These iPSC-derived macrophages were virtually indistinguishable from human blood monocyte-derived macrophages based on surface marker expression (flow cytometry), transcriptomics (RNA sequencing), and chromatin accessibility profiling. iPSC lines were additionally generated from non-human primate (chimpanzee) fibroblasts. When challenged with dengue virus, human and chimpanzee iPSC-derived macrophages show differential susceptibility to infection, thus providing a valuable resource for studying the species-tropism of viruses. We also show that blood- and iPSC-derived macrophages both restrict influenza virus at a late stage of the virus lifecycle. Collectively, our results substantiate iPSC-derived macrophages as an alternative to blood monocyte-derived macrophages for the study of virus biology. IMPORTANCE: Macrophages have complex relationships with viruses: while macrophages aid in the removal of pathogenic viruses from the body, macrophages are also manipulated by some viruses to serve as vessels for viral replication, dissemination, and long-term persistence. Here, we show that iPSC-derived macrophages are an excellent model that can be exploited in virology.


Assuntos
Vírus da Dengue , HIV-1 , Células-Tronco Pluripotentes Induzidas , Macrófagos , Modelos Biológicos , Orthomyxoviridae , Virologia , Animais , Humanos , Diferenciação Celular/genética , HIV-1/crescimento & desenvolvimento , HIV-1/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/virologia , Orthomyxoviridae/crescimento & desenvolvimento , Orthomyxoviridae/fisiologia , Pan troglodytes , Vírus da Dengue/crescimento & desenvolvimento , Vírus da Dengue/fisiologia , Fibroblastos/citologia , Monócitos/citologia , Replicação Viral , Citometria de Fluxo , Perfilação da Expressão Gênica , Montagem e Desmontagem da Cromatina , Tropismo Viral , Virologia/métodos , Biomarcadores/análise , Biomarcadores/metabolismo
14.
Cell Prolif ; 57(5): e13589, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38192172

RESUMO

Human granulosa cells in different stages are essential for maintaining normal ovarian function, and granulosa cell defect is the main cause of ovarian dysfunction. To address this problem, it is necessary to induce functional granulosa cells at different stages in vitro. In this study, we established a reprogramming method to induce early- and late-stage granulosa cells with different steroidogenic abilities. We used an AMH-fluorescence-reporter system to screen candidate factors for cellular reprogramming and generated human induced granulosa-like cells (hiGC) by overexpressing FOXL2 and NR5A1. AMH-EGFP+ hiGC resembled human cumulus cells in transcriptome profiling and secreted high levels of oestrogen and progesterone, similar to late-stage granulosa cells at antral or preovulatory stage. Moreover, we identified CD55 as a cell surface marker that can be used to isolate early-stage granulosa cells. CD55+ AMH-EGFP- hiGC secreted high levels of oestrogen but low levels of progesterone, and their transcriptome profiles were more similar to early-stage granulosa cells. More importantly, CD55+ hiGC transplantation alleviated polycystic ovary syndrome (PCOS) in a mouse model. Therefore, hiGC provides a cellular model to study the developmental program of human granulosa cells and has potential to treat PCOS.


Assuntos
Fibroblastos , Proteína Forkhead Box L2 , Células da Granulosa , Fator Esteroidogênico 1 , Feminino , Humanos , Proteína Forkhead Box L2/metabolismo , Proteína Forkhead Box L2/genética , Células da Granulosa/metabolismo , Células da Granulosa/citologia , Animais , Camundongos , Fibroblastos/metabolismo , Fibroblastos/citologia , Fator Esteroidogênico 1/metabolismo , Fator Esteroidogênico 1/genética , Progesterona/metabolismo , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Reprogramação Celular , Células Cultivadas
15.
J Biol Chem ; 300(2): 105591, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38141769

RESUMO

Long noncoding RNAs (lncRNAs) are specifically expressed in different diseases and regulate disease progression. To explore the functions of rheumatoid arthritis (RA)-specific lncRNA, we determined the lncRNA expression profile of fibroblast-like synoviocytes (FLS) obtained from patients with RA and osteoarthritis (OA) using a LncRNA microarray and identified up-regulated LncNFYB in RA as a potential therapeutic target. Using gain- and loss-of-function studies, LncNFYB was proven to promote FLS proliferation and cell cycle progress but not affect their invasion, migration, and apoptotic abilities. Further investigation discovered that LncRNA could combine with annexin A2 (ANXA2) and enhance the level of phospho-ANXA2 (Tyr24) in the plasma membrane area, which induced the activation of ERK1/2 to promote proliferation. These findings provide new insights into the biological functions of LncNFYB on modification of FLS, which may be exploited for the therapy of RA.


Assuntos
Anexina A2 , Artrite Reumatoide , Sistema de Sinalização das MAP Quinases , RNA Longo não Codificante , Sinoviócitos , Humanos , Anexina A2/genética , Anexina A2/metabolismo , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Artrite Reumatoide/fisiopatologia , Proliferação de Células/genética , Células Cultivadas , Ativação Enzimática/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/fisiopatologia , Fosforilação/genética , Ligação Proteica/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Sinoviócitos/citologia , Sinoviócitos/metabolismo
16.
Nature ; 623(7988): 792-802, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37968392

RESUMO

Optimal tissue recovery and organismal survival are achieved by spatiotemporal tuning of tissue inflammation, contraction and scar formation1. Here we identify a multipotent fibroblast progenitor marked by CD201 expression in the fascia, the deepest connective tissue layer of the skin. Using skin injury models in mice, single-cell transcriptomics and genetic lineage tracing, ablation and gene deletion models, we demonstrate that CD201+ progenitors control the pace of wound healing by generating multiple specialized cell types, from proinflammatory fibroblasts to myofibroblasts, in a spatiotemporally tuned sequence. We identified retinoic acid and hypoxia signalling as the entry checkpoints into proinflammatory and myofibroblast states. Modulating CD201+ progenitor differentiation impaired the spatiotemporal appearances of fibroblasts and chronically delayed wound healing. The discovery of proinflammatory and myofibroblast progenitors and their differentiation pathways provide a new roadmap to understand and clinically treat impaired wound healing.


Assuntos
Receptor de Proteína C Endotelial , Fáscia , Cicatrização , Animais , Camundongos , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula , Modelos Animais de Doenças , Receptor de Proteína C Endotelial/metabolismo , Fáscia/citologia , Fáscia/lesões , Fáscia/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Inflamação/metabolismo , Inflamação/patologia , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Transdução de Sinais , Análise da Expressão Gênica de Célula Única , Pele/citologia , Pele/lesões , Pele/metabolismo , Tretinoína/metabolismo
17.
Bull Exp Biol Med ; 175(5): 658-661, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37861896

RESUMO

We studied angiogenin production by human macrophages and evaluated the role of this factor in the macrophage-mediated regulation of fibroblasts. All macrophage subtypes, and especially the efferocytosis-polarized macrophages, M2(LS), actively produced angiogenin. Exogenous recombinant angiogenin dose-dependently enhanced the proliferation and differentiation of dermal fibroblasts. The addition of the angiogenin inhibitor to fibroblasts cultures suppressed the stimulating effect of exogenous angiogenin or M2(LS) conditioned media. These findings indicate the involvement of angiogenin in the macrophage-mediated paracrine regulation of skin fibroblasts.


Assuntos
Fibroblastos , Macrófagos , Ribonuclease Pancreático , Humanos , Meios de Cultivo Condicionados , Fibroblastos/citologia , Fibroblastos/metabolismo , Macrófagos/metabolismo , Ribonuclease Pancreático/metabolismo , Pele/citologia , Pele/metabolismo
18.
Sci Rep ; 13(1): 13716, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37607956

RESUMO

The enhanced availability of functional fibroblasts from precious tissue samples requires an ideal cell-culture system. Therefore, this study was designed to investigate the performance of caprine adult fibroblast cells (cadFibroblast) when cultivated in different culture media. The cadFibroblast cell lines from adult Barbari (Capra hircus) bucks were established and the effect of different media viz. DMEM/F-12 [with low-glucose (5.5 mM; DL) and high-glucose (30 mM; DH)], α-MEM [with low-glucose (5.5 mM; ML) and with high-glucose (30 mM; MH)], and fibroblast growth medium (FGM) were evaluated. Cells were then compared for growth characteristics and in-vitro dynamics through cellular morphology, proliferation, population-doubling time, double-immunocytochemistry, colony-forming units, wound healing, transwell migration, and differential expression of fibroblast-specific markers (FSP-1 and vimentin). The results of immunocytochemistry, transwell migration/invasion, and wound healing assays showed the superiority of DH over DL and other media tested. Whereas, similar effects of glucose supplementation and expression of FSP-1 were not observed in α-MEM. Transwell migration was significantly (p < 0.05) lower in FGM compared with other media tested. Overall, our results illustrate the media-dependent deviation in in-vitro dynamics and culture characteristics of cadFibroblasts that may be useful to develop strategies to cultivate these cells efficiently for research and downstream applications.


Assuntos
Meios de Cultura , Derme , Fibroblastos , Cabras , Técnicas de Cultura de Células , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/microbiologia , Meios de Cultura/química , Meios de Cultura/farmacologia , Técnicas In Vitro , Derme/citologia , Animais , Linhagem Celular , Masculino , Glucose/metabolismo , Perfilação da Expressão Gênica , Cicatrização , Ensaios de Migração Celular , Biomarcadores
19.
Nature ; 619(7971): 801-810, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37438528

RESUMO

The function of a cell is defined by its intrinsic characteristics and its niche: the tissue microenvironment in which it dwells. Here we combine single-cell and spatial transcriptomics data to discover cellular niches within eight regions of the human heart. We map cells to microanatomical locations and integrate knowledge-based and unsupervised structural annotations. We also profile the cells of the human cardiac conduction system1. The results revealed their distinctive repertoire of ion channels, G-protein-coupled receptors (GPCRs) and regulatory networks, and implicated FOXP2 in the pacemaker phenotype. We show that the sinoatrial node is compartmentalized, with a core of pacemaker cells, fibroblasts and glial cells supporting glutamatergic signalling. Using a custom CellPhoneDB.org module, we identify trans-synaptic pacemaker cell interactions with glia. We introduce a druggable target prediction tool, drug2cell, which leverages single-cell profiles and drug-target interactions to provide mechanistic insights into the chronotropic effects of drugs, including GLP-1 analogues. In the epicardium, we show enrichment of both IgG+ and IgA+ plasma cells forming immune niches that may contribute to infection defence. Overall, we provide new clarity to cardiac electro-anatomy and immunology, and our suite of computational approaches can be applied to other tissues and organs.


Assuntos
Microambiente Celular , Coração , Multiômica , Miocárdio , Humanos , Comunicação Celular , Fibroblastos/citologia , Ácido Glutâmico/metabolismo , Coração/anatomia & histologia , Coração/inervação , Canais Iônicos/metabolismo , Miocárdio/citologia , Miocárdio/imunologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Neuroglia/citologia , Pericárdio/citologia , Pericárdio/imunologia , Plasmócitos/imunologia , Receptores Acoplados a Proteínas G/metabolismo , Nó Sinoatrial/anatomia & histologia , Nó Sinoatrial/citologia , Nó Sinoatrial/fisiologia , Sistema de Condução Cardíaco/anatomia & histologia , Sistema de Condução Cardíaco/citologia , Sistema de Condução Cardíaco/metabolismo
20.
Cells ; 12(8)2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37190075

RESUMO

Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration.


Assuntos
Transdiferenciação Celular , Técnicas de Reprogramação Celular , Reprogramação Celular , Fibroblastos , Cardiopatias , Coração , Pericitos , Regeneração , Coração/fisiologia , Cardiopatias/terapia , Fibroblastos/citologia , Fibroblastos/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Pericitos/citologia , Pericitos/fisiologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA