Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.705
Filter
1.
Cells ; 13(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38727314

ABSTRACT

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.


Subject(s)
Decidua , Endometrium , Fibroblasts , RNA, Long Noncoding , Stromal Cells , Humans , Female , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Fibroblasts/metabolism , Fibroblasts/cytology , Decidua/metabolism , Decidua/cytology , Endometrium/cytology , Endometrium/metabolism , Stromal Cells/metabolism , Stromal Cells/cytology , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Pregnancy , Adult , Cell Differentiation/genetics
2.
Cells ; 13(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38786035

ABSTRACT

There are fewer investigations conducted on human primary endometrial epithelial cells (HPEECs) compared to human primary endometrial stromal cells (HPESCs). One of the main reasons is the scarcity of protocols enabling prolonged epithelial cell culture. Even though it is possible to culture HPEECs in 3D over a longer period of time, it is technically demanding. In this study, we successfully established a highly pure, stable, and long-term viable human conditionally reprogrammed endometrial epithelial cell line, designated as eCRC560. These cells stained positive for epithelial markers, estrogen and progesterone receptors, and epithelial cell-cell contacts but negative for stromal and endothelial cell markers. Estradiol (ES) reduced the abundance of ZO-1 in a time- and dose-dependent manner, in contrast to the dose-dependent increase with the progestin dienogest (DNG) when co-cultured with HPESCs. Moreover, ES significantly increased cell viability, cell migration, and invasion of the eCRC560 cells; all these effects were inhibited by pretreatment with DNG. DNG withdrawal led to a significantly disrupted monolayer of eCRC560 cells in co-culture with HPESCs, yet it markedly increased the adhesion of eCRC560 to the human mesothelial MeT-5A cells. The long-term viable eCRC560 cells are suitable for in vitro analysis of HPEECs to study the epithelial compartment of the human endometrium and endometrial pathologies.


Subject(s)
Cell Survival , Endometrium , Epithelial Cells , Estrogens , Progestins , Humans , Female , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Progestins/pharmacology , Estrogens/pharmacology , Cell Survival/drug effects , Cell Movement/drug effects , Cell Line , Estradiol/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/cytology , Coculture Techniques , Time Factors , Cell Adhesion/drug effects
3.
Stem Cell Res Ther ; 15(1): 139, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735988

ABSTRACT

The concept of "stemness" incorporates the molecular mechanisms that regulate the unlimited self-regenerative potential typical of undifferentiated primitive cells. These cells possess the unique ability to navigate the cell cycle, transitioning in and out of the quiescent G0 phase, and hold the capacity to generate diverse cell phenotypes. Stem cells, as undifferentiated precursors endow with extraordinary regenerative capabilities, exhibit a heterogeneous and tissue-specific distribution throughout the human body. The identification and characterization of distinct stem cell populations across various tissues have revolutionized our understanding of tissue homeostasis and regeneration. From the hematopoietic to the nervous and musculoskeletal systems, the presence of tissue-specific stem cells underlines the complex adaptability of multicellular organisms. Recent investigations have revealed a diverse cohort of non-hematopoietic stem cells (non-HSC), primarily within bone marrow and other stromal tissue, alongside established hematopoietic stem cells (HSC). Among these non-HSC, a rare subset exhibits pluripotent characteristics. In vitro and in vivo studies have demonstrated the remarkable differentiation potential of these putative stem cells, known by various names including multipotent adult progenitor cells (MAPC), marrow-isolated adult multilineage inducible cells (MIAMI), small blood stem cells (SBSC), very small embryonic-like stem cells (VSELs), and multilineage differentiating stress enduring cells (MUSE). The diverse nomenclatures assigned to these primitive stem cell populations may arise from different origins or varied experimental methodologies. This review aims to present a comprehensive comparison of various subpopulations of multipotent/pluripotent stem cells derived from stromal tissues. By analysing isolation techniques and surface marker expression associated with these populations, we aim to delineate the similarities and distinctions among stromal tissue-derived stem cells. Understanding the nuances of these tissue-specific stem cells is critical for unlocking their therapeutic potential and advancing regenerative medicine. The future of stem cells research should prioritize the standardization of methodologies and collaborative investigations in shared laboratory environments. This approach could mitigate variability in research outcomes and foster scientific partnerships to fully exploit the therapeutic potential of pluripotent stem cells.


Subject(s)
Multipotent Stem Cells , Pluripotent Stem Cells , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Cell Differentiation , Stromal Cells/cytology , Stromal Cells/metabolism , Animals
4.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732199

ABSTRACT

Nanofibrous materials generated through electrospinning have gained significant attention in tissue regeneration, particularly in the domain of bone reconstruction. There is high interest in designing a material resembling bone tissue, and many scientists are trying to create materials applicable to bone tissue engineering with piezoelectricity similar to bone. One of the prospective candidates is highly piezoelectric poly(vinylidene fluoride) (PVDF), which was used for fibrous scaffold formation by electrospinning. In this study, we focused on the effect of PVDF molecular weight (180,000 g/mol and 530,000 g/mol) and process parameters, such as the rotational speed of the collector, applied voltage, and solution flow rate on the properties of the final scaffold. Fourier Transform Infrared Spectroscopy allows for determining the effect of molecular weight and processing parameters on the content of the electroactive phases. It can be concluded that the higher molecular weight of the PVDF and higher collector rotational speed increase nanofibers' diameter, electroactive phase content, and piezoelectric coefficient. Various electrospinning parameters showed changes in electroactive phase content with the maximum at the applied voltage of 22 kV and flow rate of 0.8 mL/h. Moreover, the cytocompatibility of the scaffolds was confirmed in the culture of human adipose-derived stromal cells with known potential for osteogenic differentiation. Based on the results obtained, it can be concluded that PVDF scaffolds may be taken into account as a tool in bone tissue engineering and are worth further investigation.


Subject(s)
Nanofibers , Polyvinyls , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Polyvinyls/chemistry , Humans , Tissue Scaffolds/chemistry , Nanofibers/chemistry , Biocompatible Materials/chemistry , Cells, Cultured , Spectroscopy, Fourier Transform Infrared , Cell Differentiation/drug effects , Osteogenesis/drug effects , Stromal Cells/cytology , Stromal Cells/metabolism , Molecular Weight , Fluorocarbon Polymers
5.
Mech Ageing Dev ; 219: 111935, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38614143

ABSTRACT

Adipose-derived stromal cells (ADSCs) are promising stem cell sources for tissue engineering and cell-based therapy. However, long-term in vitro expansion of ADSCs impedes stemness maintenance, which is partly attributed to deprivation of their original microenvironment. Incompetent cells limit the therapeutic effects of ADSC-based clinical strategies. Therefore, reconstructing a more physiologically and physically relevant niche is an ideal strategy to address this issue and therefore facilitates the extensive application of ADSCs. Here, we transplanted separated ADSCs into local subcutaneous adipose tissues of nude mice as an in vivo cell culture model. We found that transplanted ADSCs maintained their primitive morphology and showed improved proliferation and delayed senescence compared to those of cells cultured in an incubator. Significantly increased expression of stemness-related markers and multilineage differentiation abilities were further observed in in vivo cultured ADSCs. Finally, sequencing revealed that genes whose expression differed between ADSCs obtained under in vivo and in vitro conditions were mainly located in the extracellular matrix and extracellular space and that these genes participate in regulating transcription and protein synthesis. Moreover, we found that an Egr1 signaling pathway might exert a crucial impact on controlling stemness properties. Our findings might collectively pave the way for ADSC-based applications.


Subject(s)
Cell Differentiation , Cell Proliferation , Animals , Rats , Cell Differentiation/physiology , Mice , Adipose Tissue/cytology , Adipose Tissue/metabolism , Mice, Nude , Stromal Cells/metabolism , Stromal Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Early Growth Response Protein 1/metabolism , Male , Cells, Cultured
6.
Stem Cell Res ; 77: 103417, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608355

ABSTRACT

The pro-angiogenic abilities of adipose-derived stromal cells (ASCs) make them attractive candidates for cellular therapy, especially for ischemic disease indications. However, details regarding the underlying mechanisms remain elusive. Therefore, this study aimed to investigate paracrine and juxtacrine abilities of ASCs in angiogenesis triple cell co-cultures by detailed image analysis of the vascular-like structures. Fibroblast-endothelial cell co-cultures were established, and ASCs were added directly or indirectly through inserts. The cultures were treated with antibodies or subjected to analyses using ELISA and RT2 PCR Arrays. The model consistently generated vascular-like structures. ASCs increased the total branch lengths equally well in paracrine and juxtacrine conditions, by increasing the number of branches and average branch lengths (ABL). In contrast, addition of VEGF to the model increased the number of branches, but not the ABL. Still, ASCs increased the VEGF levels in supernatants of paracrine and juxtacrine co-cultures, and anti-VEGF treatment decreased the sprouting. ASCs themselves up-regulated collagen type V in response to paracrine signals from the co-cultures. The results suggest that ASCs initiate sprouting through secretion of several paracrine factors, among which VEGF is identified, but VEGF alone does not recapitulate the paracrine actions of ASCs. By employing neutralizing antibodies and dismantling common model outputs using image analysis, the triple cell co-culture is an attractive tool for discovery of the paracrine factors in ASCs' secretome which act in concert with VEGF to improve angiogenesis.


Subject(s)
Adipose Tissue , Coculture Techniques , Neovascularization, Physiologic , Paracrine Communication , Stromal Cells , Humans , Stromal Cells/metabolism , Stromal Cells/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/cytology , Angiogenesis
7.
Soft Matter ; 20(16): 3483-3498, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38587658

ABSTRACT

A breast-cancer tumor develops within a stroma, a tissue where a complex extracellular matrix surrounds cells, mediating the cancer progression through biomechanical and -chemical cues. Current materials partially mimic the stromal matrix in 3D cell cultures but methods for measuring the mechanical properties of the matrix at cell-relevant-length scales and stromal-stiffness levels are lacking. Here, to address this gap, we developed a characterization approach that employs probe-based microrheometry and Bayesian modeling to quantify length-scale-dependent mechanics and mechanical heterogeneity as in the stromal matrix. We examined the interpenetrating network (IPN) composed of alginate scaffolds (for adjusting mechanics) and type-1 collagen (a stromal-matrix constituent). We analyzed viscoelasticity: absolute-shear moduli (stiffness/elasticity) and phase angles (viscous and elastic characteristics). We determined the relationship between microrheometry and rheometry information. Microrheometry reveals lower stiffness at cell-relevant scales, compared to macroscale rheometry, with dependency on the length scale (10 to 100 µm). These data show increasing IPN stiffness with crosslinking until saturation (≃15 mM of Ca2+). Furthermore, we report that IPN stiffness can be adjusted by modulating collagen concentration and interconnectivity (by polymerization temperature). The IPNs are heterogeneous structurally (in SEM) and mechanically. Interestingly, increased alginate crosslinking changes IPN heterogeneity in stiffness but not in phase angle, until the saturation. In contrast, such changes are undetectable in alginate scaffolds. Our nonlinear viscoelasticity analysis at tumor-cell-exerted strains shows that only the softer IPNs stiffen with strain, like the stromal-collagen constituent. In summary, our approach can quantify the stromal-matrix-related viscoelasticity and is likely applicable to other materials in 3D culture.


Subject(s)
Alginates , Extracellular Matrix , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Alginates/chemistry , Cell Culture Techniques, Three Dimensional , Viscosity , Stromal Cells/cytology , Stromal Cells/metabolism , Elasticity , Tissue Scaffolds/chemistry , Collagen Type I/chemistry , Collagen Type I/metabolism , Biomechanical Phenomena , Rheology , Models, Biological , Bayes Theorem
8.
Adv Healthc Mater ; 13(13): e2303674, 2024 May.
Article in English | MEDLINE | ID: mdl-38315148

ABSTRACT

Intrauterine adhesion (IUA) stands as a prevalent medical condition characterized by endometrial fibrosis and scar tissue formation within the uterine cavity, resulting in infertility and, in severe cases, recurrent miscarriages. Cell therapy, especially with stem cells, offers an alternative to surgery, but concerns about uncontrolled differentiation and tumorigenicity limit its use. Exosomes, more stable and immunogenicity-reduced than parent cells, have emerged as a promising avenue for IUA treatment. In this study, a novel approach has been proposed wherein exosomes originating from decidual stromal cells (DSCs) are encapsulated within sodium alginate hydrogel (SAH) scaffolds to repair endometrial damage and restore fertility in a mouse IUA model. Current results demonstrate that in situ injection of DSC-derived exosomes (DSC-exos)/SAH into the uterine cavity has the capability to induce uterine angiogenesis, initiate mesenchymal-to-epithelial transformation (MET), facilitate collagen fiber remodeling and dissolution, promote endometrial regeneration, enhance endometrial receptivity, and contribute to the recovery of fertility. RNA sequencing and advanced bioinformatics analysis reveal miRNA enrichment in exosomes, potentially supporting endometrial repair. This finding elucidates how DSC-exos/SAH mechanistically fosters collagen ablation, endometrium regeneration, and fertility recovery, holding the potential to introduce a novel IUA treatment and offering invaluable insights into the realm of regenerative medicine.


Subject(s)
Alginates , Endometrium , Exosomes , Hydrogels , Regeneration , Stromal Cells , Female , Alginates/chemistry , Exosomes/metabolism , Exosomes/chemistry , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Endometrium/cytology , Endometrium/metabolism , Mice , Regeneration/drug effects , Stromal Cells/metabolism , Stromal Cells/cytology , Decidua/cytology , Decidua/metabolism , Fertility/physiology , MicroRNAs/metabolism , MicroRNAs/genetics , Humans , Tissue Adhesions/metabolism
9.
World J Microbiol Biotechnol ; 39(10): 276, 2023 Aug 12.
Article in English | MEDLINE | ID: mdl-37567959

ABSTRACT

The increasing number of life-threatening infections caused by persister bacteria is associated with various issues, including antimicrobial resistance and biofilm formation. Infections due to persister cells are often difficult to suppress without the use of last-resort antibiotics. Throughout the world, bacterial persistence and resistance create an unmet clinical demand for the exploration of newly introduced therapeutic approaches. Mesenchymal stem / stromal cells (MSCs) have an antimicrobial activity to protect against bacterial infections, including those caused by bacterial persisters. MSCs have substantial potential to secrete antimicrobial peptides (AMPs), including cathelicidin, beta-defensins, lipocalin-2, hepcidin, indoleamine 2,3-dioxygenase (IDO), cysteine proteases, and inducible nitric oxide synthases (iNOS). MSCs possess the potential to contribute to innate immunity by regulating the immune response. Recently, MSCs and their secreted components have been reported to improve antimicrobial activity. Bactericidal activity by MSCs and their secretomes has been shown to be mediated in part by the secretion of AMPs. Even though they were discovered more than 80 years ago, therapeutic options for persisters are restricted, and there is an urgent need for alternative treatment regimens. Hence, this review intends to critically assess the current literature on the effects of MSCs and their secretomes on persister bacteria. MSCs and their secretome-based therapies could be preferred as an up-and-coming approach to reinforce the antimicrobial efficiency in persister infections.


Subject(s)
Bacterial Infections , Cell- and Tissue-Based Therapy , Mesenchymal Stem Cells , Secretome , Stromal Cells , Antimicrobial Peptides/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Bacterial Infections/microbiology , Bacterial Infections/therapy , Humans , Animals , Stromal Cells/cytology , Stromal Cells/metabolism
10.
J Biol Chem ; 299(6): 104805, 2023 06.
Article in English | MEDLINE | ID: mdl-37172728

ABSTRACT

Bone development starts with condensations of undifferentiated mesenchymal cells that set a framework for future bones within the primordium. In the endochondral pathway, mesenchymal cells inside the condensation differentiate into chondrocytes and perichondrial cells in a SOX9-dependent mechanism. However, the identity of mesenchymal cells outside the condensation and how they participate in developing bones remain undefined. Here we show that mesenchymal cells surrounding the condensation contribute to both cartilage and perichondrium, robustly generating chondrocytes, osteoblasts, and marrow stromal cells in developing bones. Single-cell RNA-seq analysis of Prrx1-cre-marked limb bud mesenchymal cells at E11.5 reveals that Notch effector Hes1 is expressed in a mutually exclusive manner with Sox9 that is expressed in pre-cartilaginous condensations. Analysis of a Notch signaling reporter CBF1:H2B-Venus reveals that peri-condensation mesenchymal cells are active for Notch signaling. In vivo lineage-tracing analysis using Hes1-creER identifies that Hes1+ early mesenchymal cells surrounding the SOX9+ condensation at E10.5 contribute to both cartilage and perichondrium at E13.5, subsequently becoming growth plate chondrocytes, osteoblasts of trabecular and cortical bones, and marrow stromal cells in postnatal bones. In contrast, Hes1+ cells in the perichondrium at E12.5 or E14.5 do not generate chondrocytes within cartilage, contributing to osteoblasts and marrow stromal cells only through the perichondrial route. Therefore, Hes1+ peri-condensation mesenchymal cells give rise to cells of the skeletal lineage through cartilage-dependent and independent pathways, supporting the theory that early mesenchymal cells outside the condensation also play important roles in early bone development.


Subject(s)
Bone Development , Bone and Bones , Cartilage , Cell Differentiation , Cell Lineage , Chondrocytes , Mesenchymal Stem Cells , Transcription Factor HES-1 , Animals , Mice , Bone and Bones/cytology , Cartilage/cytology , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factor HES-1/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Receptors, Notch/metabolism
11.
Proc Natl Acad Sci U S A ; 119(38): e2200252119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36095212

ABSTRACT

In humans, the uterus undergoes a dramatic transformation to form an endometrial stroma-derived secretory tissue, termed decidua, during early pregnancy. The decidua secretes various factors that act in an autocrine/paracrine manner to promote stromal differentiation, facilitate maternal angiogenesis, and influence trophoblast differentiation and development, which are critical for the formation of a functional placenta. Here, we investigated the mechanisms by which decidual cells communicate with each other and with other cell types within the uterine milieu. We discovered that primary human endometrial stromal cells (HESCs) secrete extracellular vesicles (EVs) during decidualization and that this process is controlled by a conserved HIF2α-RAB27B pathway. Mass spectrometry revealed that the decidual EVs harbor a variety of protein cargo, including cell signaling molecules, growth modulators, metabolic regulators, and factors controlling endothelial cell expansion and remodeling. We tested the hypothesis that EVs secreted by the decidual cells mediate functional communications between various cell types within the uterus. We demonstrated that the internalization of EVs, specifically those carrying the glucose transporter 1 (GLUT1), promotes glucose uptake in recipient HESCs, supporting and advancing the decidualization program. Additionally, delivery of HESC-derived EVs into human endothelial cells stimulated their proliferation and led to enhanced vascular network formation. Strikingly, stromal EVs also promoted the differentiation of trophoblast stem cells into the extravillous trophoblast lineage. Collectively, these findings provide a deeper understanding of the pleiotropic roles played by EVs secreted by the decidual cells to ensure coordination of endometrial differentiation and angiogenesis with trophoblast function during the progressive phases of decidualization and placentation.


Subject(s)
Decidua , Extracellular Vesicles , Trophoblasts , Cell Differentiation , Decidua/cytology , Decidua/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Extracellular Vesicles/physiology , Female , Humans , Neovascularization, Physiologic , Pregnancy , Stromal Cells/cytology , Stromal Cells/physiology , Trophoblasts/cytology , Trophoblasts/physiology
12.
Nature ; 608(7924): 733-740, 2022 08.
Article in English | MEDLINE | ID: mdl-35978187

ABSTRACT

Single-cell transcriptomics (scRNA-seq) has greatly advanced our ability to characterize cellular heterogeneity1. However, scRNA-seq requires lysing cells, which impedes further molecular or functional analyses on the same cells. Here, we established Live-seq, a single-cell transcriptome profiling approach that preserves cell viability during RNA extraction using fluidic force microscopy2,3, thus allowing to couple a cell's ground-state transcriptome to its downstream molecular or phenotypic behaviour. To benchmark Live-seq, we used cell growth, functional responses and whole-cell transcriptome read-outs to demonstrate that Live-seq can accurately stratify diverse cell types and states without inducing major cellular perturbations. As a proof of concept, we show that Live-seq can be used to directly map a cell's trajectory by sequentially profiling the transcriptomes of individual macrophages before and after lipopolysaccharide (LPS) stimulation, and of adipose stromal cells pre- and post-differentiation. In addition, we demonstrate that Live-seq can function as a transcriptomic recorder by preregistering the transcriptomes of individual macrophages that were subsequently monitored by time-lapse imaging after LPS exposure. This enabled the unsupervised, genome-wide ranking of genes on the basis of their ability to affect macrophage LPS response heterogeneity, revealing basal Nfkbia expression level and cell cycle state as important phenotypic determinants, which we experimentally validated. Thus, Live-seq can address a broad range of biological questions by transforming scRNA-seq from an end-point to a temporal analysis approach.


Subject(s)
Cell Survival , Gene Expression Profiling , Macrophages , RNA-Seq , Single-Cell Analysis , Transcriptome , Adipose Tissue/cytology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Differentiation , Gene Expression Profiling/methods , Gene Expression Profiling/standards , Genome/drug effects , Genome/genetics , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , NF-KappaB Inhibitor alpha/genetics , Organ Specificity , Phenotype , RNA/genetics , RNA/isolation & purification , RNA-Seq/methods , RNA-Seq/standards , Reproducibility of Results , Sequence Analysis, RNA/methods , Sequence Analysis, RNA/standards , Single-Cell Analysis/methods , Stromal Cells/cytology , Stromal Cells/metabolism , Time Factors , Transcriptome/genetics
13.
Cell Biochem Funct ; 40(5): 439-450, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35707856

ABSTRACT

Regular soft tissue healing relies on the well-organized interaction of different stromal cell types with endothelial cells. However, spatiotemporal conditions might provoke high densities of one special stromal cell type, potentially leading to impaired healing. Detailed knowledge of the functions of rivaling stromal cell types aiming for tissue contraction and stabilization as well as vascular support is mandatory. By the application of an in vitro approach comprising the evaluation of cell proliferation, cell morphology, myofibroblastoid differentiation, and cytokine release, we verified a density-dependent modulation of these functions among juvenile and adult fibroblasts, pericytes, and adipose-derived stem cells during their interaction with microvascular endothelial cells in cocultures. Results indicate that juvenile fibroblasts rather support angiogenesis via paracrine regulation at the early stage of healing, a role potentially compromised in adult fibroblasts. In contrast, pericytes showed a more versatile character aiming at angiogenesis, vessel stabilization, and tissue contraction. Such a universal character was even more pronounced among adipose-derived stem cells. The explicit knowledge of the characteristic functions of stromal cell types is a prerequisite for the development of new analytical and therapeutic approaches for impaired soft tissue healing. The present study delivers new considerations concerning the roles of rivaling stromal cell types within a granulation tissue, pointing to extraordinary properties of pericytes and adipose-derived stem cells.


Subject(s)
Endothelial Cells , Stromal Cells , Wound Healing , Adipose Tissue/cytology , Cell Count , Endothelial Cells/cytology , Fibroblasts/cytology , Humans , Neovascularization, Pathologic , Pericytes/cytology , Stem Cells/cytology , Stromal Cells/cytology
14.
PLoS One ; 17(2): e0263141, 2022.
Article in English | MEDLINE | ID: mdl-35120168

ABSTRACT

Corneal grafts are the imperative clinical treatment for canine corneal blindness. To serve the growing demand, this study aimed to generate tissue-engineered canine cornea in part of the corneal epithelium and underlying stroma based on canine limbal epithelial stem cells (cLESCs) seeded silk fibroin/gelatin (SF/G) film and canine corneal stromal stem cells (cCSSCs) seeded SF/G scaffold, respectively. Both cell types were successfully isolated by collagenase I. SF/G corneal films and stromal scaffolds served as the prospective substrates for cLESCs and cCSSCs by promoting cell adhesion, cell viability, and cell proliferation. The results revealed the upregulation of tumor protein P63 (P63) and ATP-binding cassette super-family G member 2 (Abcg2) of cLESCs as well as Keratocan (Kera), Lumican (Lum), aldehyde dehydrogenase 3 family member A1 (Aldh3a1) and Aquaporin 1 (Aqp1) of differentiated keratocytes. Moreover, immunohistochemistry illustrated the positive staining of tumor protein P63 (P63), aldehyde dehydrogenase 3 family member A1 (Aldh3a1), lumican (Lum) and collagen I (Col-I), which are considerable for native cornea. This study manifested a feasible platform to construct tissue-engineered canine cornea for functional grafts and positively contributed to the body of knowledge related to canine corneal stem cells.


Subject(s)
Biocompatible Materials/chemistry , Epithelium, Corneal/pathology , Regeneration , Stem Cells/cytology , Stromal Cells/cytology , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Aquaporin 1/metabolism , Cell Proliferation , Collagen Type I/metabolism , Corneal Transplantation , Dogs , Eye Proteins/metabolism , Fibroblasts/cytology , Fibroins/chemistry , Gelatin/chemistry , Genes, Tumor Suppressor , Immunohistochemistry , In Vitro Techniques , Lumican/metabolism , Mice , Tensile Strength , Tissue Engineering , Tissue Scaffolds
15.
Sci Rep ; 12(1): 1775, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110646

ABSTRACT

Human adipose tissue is a rich source of adipose-derived stem cells (ASCs) and vascular endothelial progenitor cells (EPCs). However, no standardized method has been established for the isolation and purification of adipose-derived EPCs (AEPCs). The aim of this study was to establish a method for the isolation and purification of AEPCs. The stromal vascular fraction (SVF) was extracted from human lipoaspirates, and the CD45-CD31+ fraction of the SVF was collected by magnetic-activated cell sorting (MACS). The CD45-CD31+ fraction was cultured for 4.5 days, followed by a second MACS separation to collect the CD31+ fraction. Purified AEPCs were expanded without being overwhelmed by proliferating ASCs, indicating that a high level (> 95%) of AEPC purification is a key factor for their successful isolation and expansion. AEPCs exhibited typical endothelial markers, including CD31, von Willebrand factor, and the isolectin-B4 binding capacity. AEPCs formed colonies, comparable to cultured human umbilical vein endothelial cells (HUVECs). Both AEPCs and HUVECs formed capillary-like networks in the tube formation assay, with no significant difference in network lengths. We are the first to establish a purification and expansion method to isolate these cells. Because adipose tissue is a clinically accessible and abundant tissue, AEPCs may have potential advantages as a therapeutic tool for regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Biomarkers/metabolism , Endothelial Progenitor Cells/cytology , Regenerative Medicine , Stromal Cells/cytology , Adipose Tissue/metabolism , Adult , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Female , Humans , Male , Middle Aged , Stromal Cells/metabolism
16.
Pharm Biol ; 60(1): 501-508, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35188840

ABSTRACT

CONTEXT: The osteogenic potential of the human dental pulp stromal cells (hDPSCs) was reduced in the state of oxidative stress. Resveratrol (RSV) possesses numerous biological properties, including osteogenic potential, growth-promoting and antioxidant activities. OBJECTIVE: This study investigates the osteogenic potential of RSV by activating the Sirt1/Nrf2 pathway on oxidatively stressed hDPSCs and old mice. MATERIALS AND METHODS: The hDPSCs were subjected to reactive oxygen species (ROS) fluorescence staining, cell proliferation assay, ROS activity assay, superoxide dismutase (SOD) enzyme activity, the glutathione (GSH) concentration assay, alkaline phosphatase staining, real-time polymerase chain reaction (RT-PCR) and Sirt1 immunofluorescence labelling to assess the antioxidant stress and osteogenic ability of RSV. Forty female Kunming mice were divided into Old, Old-RSV, Young and Young-RSV groups to assess the repair of calvarial defects of 0.2 mL RSV of 20 mg/kg/d for seven days by injecting intraperitoneally at 4 weeks after surgery using micro-computed tomography, nonlinear optical microscope and immunohistochemical analysis. RESULTS: RSV abates oxidative stress by alleviating the proliferation, mitigating the ROS activity, increasing the SOD enzyme activity and ameliorating the GSH concentration (RSV IC50 in hDPSCs is 67.65 ± 9.86). The antioxidative stress and osteogenic capabilities of RSV were confirmed by the up-regulated gene expression of SOD1, xCT, RUNX2 and OCN, as well as Sirt1/Nrf2. The collagen, bone matrix formation and Sirt1 expression, are significantly increased after RSV treatment in mice. DISCUSSION AND CONCLUSIONS: For elderly or patients with oxidative stress physiological states such as hypertension, heart disease, diabetes, etc., RSV may potentially improve bone augmentation surgery in regenerative medicine.


Subject(s)
Osteogenesis/drug effects , Oxidative Stress/drug effects , Resveratrol/pharmacology , Stromal Cells/drug effects , Age Factors , Animals , Animals, Outbred Strains , Cell Proliferation/drug effects , Cells, Cultured , Dental Pulp/cytology , Female , Humans , Mice , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Stromal Cells/cytology , Superoxide Dismutase/metabolism
17.
Sci Rep ; 12(1): 2414, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35165317

ABSTRACT

Pediatric patients with Osteogenesis Imperfecta (OI), a heritable connective tissue disorder, frequently suffer from long bone deformations. Surgical correction often results in bone non-unions, necessitating revision surgery with autogenous bone grafting using bone-marrow-derived stem cells (BM-SC) to regenerate bone. BM-SC harvest is generally invasive and limited in supply; thus, adipose tissue's stromal vascular fraction (SVF) has been introduced as an alternative stem cell reservoir. To elucidate if OI patients' surgical site dissected adipose tissue could be used as autologous bone graft in future, we investigated whether the underlying genetic condition alters SVF's cell populations and in vitro differentiation capacity. After optimizing SVF isolation, we demonstrate successful isolation of SVF of pediatric OI patients and non-OI controls. The number of viable cells was comparable between OI and controls, with about 450,000 per gram tissue. Age, sex, type of OI, disease-causing collagen mutation, or anatomical site of harvest did not affect cell outcome. Further, SVF-containing cell populations were similar between OI and controls, and all isolated SVF's demonstrated chondrogenic, adipogenic, and osteogenic differentiation capacity in vitro. These results indicate that SVF from pediatric OI patients could be used as a source of stem cells for autologous stem cell therapy in OI.


Subject(s)
Adipose Tissue/cytology , Osteogenesis Imperfecta/physiopathology , Stromal Cells/cytology , Adipogenesis , Adipose Tissue/metabolism , Adolescent , Child , Child, Preschool , Chondrogenesis , Collagen/genetics , Collagen/metabolism , Female , Humans , Male , Mutation , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Stromal Cells/metabolism , Stromal Vascular Fraction/metabolism , Young Adult
18.
Int J Cardiol ; 352: 104-114, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35074490

ABSTRACT

OBJECTIVE: To explore the mechanism of CircANKRD36 regulating cell heterogeneity and endothelial mesenchymal transition in aortic valve stromal cells by regulating miR-599 and TGF-ß signaling pathway. METHODS: Human tissue specimens were divided into Control group (n = 25) and CAVD group (n = 25). The mRNA expressions of CircANKRD36 and miR-599 in tissue samples were analyzed by qRT-PCR. Western blot was used to analyze the protein expression of osteogenic differentiation related factors induced by OM.The expressions of ALP, osteocalcin, osteopontin, Runx2 and Cadherin11 were detected by Western blot. RESULTS: The expression of CircANKRD36mRNA in CAVD tissue was lower than that in Control tissue (P < 0.05), and the expression of miR-599mRNA in CAVD tissue was higher than that in Control tissue (P < 0.05). CircANKRD36 was negatively correlated with ALP, osteocalcin, osteopontin, Runx2, Cadherin11 expression level after OM induced osteogenic differentiation. The expression level of miR-599 was positively correlated with ALP, osteocalcin, osteopontin, Runx2 and Cadherin11 after OM induced osteogenic differentiation.The expression of ALP, osteocalcin, osteopontin, Runx2 and Cadherin11 protein in circ+miR-599 group was lower than that in circ+miR-NC group (P < 0.05). Compared with Vector+miR-NC group, the protein expressions of TGF-ß1, TGF-ß2 and SMAD4 in circ+miR-NC group decreased (P < 0.05), while the protein expressions of TGF-ß1, TGF-ß2 and SMAD4 in circ+miR-599 group increased (P < 0.05). CONCLUSION: CircANKRD36 can inhibit the expression of miR-599 and the activation of TGF-ß signaling pathway, thus inhibiting the expression of differentiation-related factors of VIC osteogenesis and the formation of calcified nodules. Therefore, circANKRD36-miR-599-TGF-ß axis can be a new theoretical basis for treating CAVD.


Subject(s)
MicroRNAs , Nuclear Proteins , Osteogenesis , Stromal Cells , Aortic Diseases , Aortic Valve/cytology , Aortic Valve/metabolism , Calcinosis , Cell Differentiation , Cells, Cultured , Humans , Nuclear Proteins/genetics , Osteogenesis/genetics , RNA, Circular , Signal Transduction/genetics , Stromal Cells/cytology , Stromal Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
PLoS Genet ; 18(1): e1010018, 2022 01.
Article in English | MEDLINE | ID: mdl-35025868

ABSTRACT

Approximately 75% of failed pregnancies are considered to be due to embryo implantation failure or defects. Nevertheless, the explicit signaling mechanisms governing this process have not yet been elucidated. Here, we found that conditional deletion of the Shp2 gene in mouse uterine stromal cells deferred embryo implantation and inhibited the decidualization of stromal cells, which led to embryonic developmental delay and to the death of numerous embryos mid-gestation, ultimately reducing female fertility. The absence of Shp2 in stromal cells increased the proliferation of endometrial epithelial cells, thereby disturbing endometrial epithelial remodeling. However, Shp2 deletion impaired the proliferation and polyploidization of stromal cells, which are distinct characteristics of decidualization. In human endometrial stromal cells (hESCs), Shp2 expression gradually increased during the decidualization process. Knockout of Shp2 blocked the decidual differentiation of hESCs, while Shp2 overexpression had the opposite effect. Shp2 knockout inhibited the proliferation of hESCs during decidualization. Whole gene expression profiling analysis of hESCs during the decidualization process showed that Shp2 deficiency disrupted many signaling transduction pathways and gene expression. Analyses of hESCs and mouse uterine tissues confirmed that the signaling pathways extracellular regulated protein kinases (ERK), protein kinase B (AKT), signal transducer and activator of transcription 3 (STAT3) and their downstream transcription factors CCAAT/enhancer binding protein ß (C/EBPß) and Forkhead box transcription factor O1 (FOXO-1) were involved in the Shp2 regulation of decidualization. In summary, these results demonstrate that Shp2 plays a crucial role in stromal decidualization by mediating and coordinating multiple signaling pathways in uterine stromal cells. Our discovery possibly provides a novel key regulator of embryo implantation and novel therapeutic target for pregnancy failure.


Subject(s)
Decidua/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Uterus/cytology , Animals , Cell Line , Cell Proliferation , Embryo Implantation , Female , Gene Deletion , Gene Expression Profiling , Humans , Mice , Pregnancy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , Uterus/metabolism
20.
PLoS One ; 17(1): e0261498, 2022.
Article in English | MEDLINE | ID: mdl-35025920

ABSTRACT

BACKGROUND: A paracrine mechanism is thought to mediate the proangiogenic capacity of adipose-derived stromal/stem cells (ASCs). However, the precise mechanism by which ASCs promote the formation of blood vessels by endothelial progenitor cells (EPCs) is unclear. METHODS: The EPCs-ASCs cocultures prepared in different ratios were subjected to tube formations assay to verify whether ASCs could directly participate in the tube genesis. The supernatant from cultured ASCs was used to stimulate EPCs to evaluate the effects on the angiogenic property of EPCs, as well as capacity for migration and invasion. A coculture model with transwell chamber were used to explore the regulation of angiogenesis markers expression in EPCs by ASCs. We then mixed ASCs with EPCs and transplanted them with adipose tissue into nude mice to evaluate the effects on angiogenesis in adipose tissue grafts. RESULTS: In the EPCs-ASCs cocultures, the tube formation was significantly decreased as the relative abundance of ASCs increased, while the ASCs was found to migrate and integrated into the agglomerates formed by EPCs. The supernatant from ASCs cultures promoted the migration and invasion of EPCs and the ability to form capillary-like structures. The expression of multiple angiogenesis markers in EPCs were significantly increased when cocultured with ASCs. In vivo, ASCs combined with EPC promoted vascularization in the fat transplant. Immunofluorescence straining of Edu and CD31 indicated that the Edu labeled EPC did not directly participate in the vascularization inside the fat tissue. CONCLUSIONS: ADSC can participate in the tube formation of EPC although it cannot form canonical capillary structures. Meanwhile, Soluble factors secreted by ASCs promotes the angiogenic potential of EPCs. ASCs paracrine signaling appears to promote angiogenesis by increasing the migration and invasion of EPCs and simultaneously upregulating the expression of angiogenesis markers in EPCs. The results of in vivo experiments showed that ASCs combined with EPCs significantly promote the formation of blood vessels in the fat implant. Remarkably, EPCs may promote angiogenesis by paracrine regulation of endogenous endothelial cells (ECs) rather than direct participation in the formation of blood vessels.


Subject(s)
Endothelial Progenitor Cells/transplantation , Graft Survival/physiology , Neovascularization, Physiologic/physiology , Stromal Cells/transplantation , Adipose Tissue/cytology , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Cell Culture Techniques , Cell Movement , Cells, Cultured , Coculture Techniques , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Mice , Mice, Nude , Paracrine Communication/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rabbits , Stromal Cells/cytology , Stromal Cells/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...