Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biomaterials ; 306: 122471, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38377846

ABSTRACT

Allogeneic stem-cell based regenerative medicine is a promising approach for bone defect repair. The use of chondrogenically differentiated human marrow stromal cells (MSCs) has been shown to lead to bone formation by endochondral ossification in immunodeficient pre-clinical models. However, an insight into the interactions between the allogeneic immune system and the human MSC-derived bone grafts has not been fully achieved yet. The choice of a potent source of MSCs isolated from pediatric donors with consistent differentiation and high proliferation abilities, as well as low immunogenicity, could increase the chance of success for bone allografts. In this study, we employed an immunodeficient animal model humanised with allogeneic immune cells to study the immune responses towards chondrogenically differentiated human pediatric MSCs (ch-pMSCs). We show that ch-differentiated pMSCs remained non-immunogenic to allogeneic CD4 and CD8 T cells in an in vitro co-culture model. After subcutaneous implantation in mice, ch-pMSC-derived grafts were able to initiate bone mineralisation in the presence of an allogeneic immune system for 3 weeks without the onset of immune responses. Re-exposing the splenocytes of the humanised animals to pMSCs did not trigger further T cell proliferation, suggesting an absence of secondary immune responses. Moreover, ch-pMSCs generated mature bone after 8 weeks of implantation that persisted for up to 6 more weeks in the presence of an allogeneic immune system. These data collectively show that human allogeneic chondrogenically differentiated pediatric MSCs might be a safe and potent option for bone defect repair in the tissue engineering and regenerative medicine setting.


Subject(s)
Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mice , Animals , Child , Osteogenesis , Bone Marrow , Stromal Cells , Cell Differentiation , Bone Marrow Cells , Cells, Cultured
2.
Cells ; 12(8)2023 04 20.
Article in English | MEDLINE | ID: mdl-37190110

ABSTRACT

Tissue engineering bone via endochondral ossification requires the generation of a cartilage template which undergoes vascularisation and remodelling. While this is a promising route for bone repair, achieving effective cartilage vascularisation remains a challenge. Here, we investigated how mineralisation of tissue-engineered cartilage affects its pro-angiogenic potential. To generate in vitro mineralised cartilage, human mesenchymal stromal cell (hMSC)-derived chondrogenic pellets were treated with ß-glycerophosphate (BGP). After optimising this approach, we characterised the changes in matrix components and pro-angiogenic factors by gene expression analysis, histology and ELISA. Human umbilical vein endothelial cells (HUVECs) were exposed to pellet-derived conditioned media, and migration, proliferation and tube formation were assessed. We established a reliable strategy to induce in vitro cartilage mineralisation, whereby hMSC pellets are chondrogenically primed with TGF-ß for 2 weeks and BGP is added from week 2 of culture. Cartilage mineralisation determines loss of glycosaminoglycans, reduced expression but not protein abundance of collagen II and X, and decreased VEGFA production. Finally, the conditioned medium from mineralised pellets showed a reduced ability to stimulate endothelial cell migration, proliferation and tube formation. The pro-angiogenic potential of transient cartilage is thus stage-dependent, and this aspect must be carefully considered in the design of bone tissue engineering strategies.


Subject(s)
Cartilage , Tissue Engineering , Humans , Tissue Engineering/methods , Cartilage/metabolism , Calcification, Physiologic , Human Umbilical Vein Endothelial Cells , Cell Proliferation
3.
Front Immunol ; 12: 715267, 2021.
Article in English | MEDLINE | ID: mdl-34659205

ABSTRACT

Tissue engineering approaches using progenitor cells such as mesenchymal stromal cells (MSCs) represent a promising strategy to regenerate bone. Previous work has demonstrated the potential of chondrogenically primed human MSCs to recapitulate the process of endochondral ossification and form mature bone in vivo, using immunodeficient xenogeneic models. To further the translation of such MSC-based approaches, additional investigation is required to understand the impact of interactions between human MSC constructs and host immune cells upon the success of MSC-mediated bone formation. Although human MSCs are considered hypoimmunogenic, the potential of chondrogenically primed human MSCs to induce immunogenic responses in vivo, as well as the efficacy of MSC-mediated ectopic bone formation in the presence of fully competent immune system, requires further elucidation. Therefore, the aim of this study was to investigate the capacity of chondrogenically primed human MSC constructs to persist and undergo the process of endochondral ossification in an immune competent xenogeneic model. Chondrogenically differentiated human MSC pellets were subcutaneously implanted to wild-type BALB/c mice and retrieved at 2 and 12 weeks post-implantation. The percentages of CD4+ and CD8+ T cells, B cells, and classical/non-classical monocyte subsets were not altered in the peripheral blood of mice that received chondrogenic MSC constructs compared to sham-operated controls at 2 weeks post-surgery. However, MSC-implanted mice had significantly higher levels of serum total IgG compared to sham-operated mice at this timepoint. Flow cytometric analysis of retrieved MSC constructs identified the presence of T cells and macrophages at 2 and 12 weeks post-implantation, with low levels of immune cell infiltration to implanted MSC constructs detected by CD45 and CD3 immunohistochemical staining. Despite the presence of immune cells in the tissue, MSC constructs persisted in vivo and were not degraded/resorbed. Furthermore, constructs became mineralised, with longitudinal micro-computed tomography imaging revealing an increase in mineralised tissue volume from 4 weeks post-implantation until the experimental endpoint at 12 weeks. These findings indicate that chondrogenically differentiated human MSC pellets can persist and undergo early stages of endochondral ossification following subcutaneous implantation in an immunocompetent xenogeneic model. This scaffold-free model may be further extrapolated to provide mechanistic insight to osteoimmunological processes regulating bone regeneration and homeostasis.


Subject(s)
Calcification, Physiologic , Cell Differentiation , Chondrogenesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Biomarkers , Bone Regeneration , Cell Differentiation/genetics , Cells, Cultured , Chondrogenesis/genetics , Humans , Immunity , Mice , Models, Animal , Monocytes/immunology , Monocytes/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tissue Engineering , X-Ray Microtomography
4.
J Control Release ; 333: 28-40, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33741386

ABSTRACT

Antisense oligonucleotides (ASOs) carry an enormous therapeutic potential in different research areas, however, the lack of appropriate carriers for their delivery to the target tissues is hampering their clinical translation. The present study investigates the application of novel biomimetic nano-vesicles, Nano-Ghosts (NGs), for the delivery of ASOs to human mesenchymal stem cells (MSCs), using a microRNA inhibitor (antimiR) against miR-221 as proof-of-concept. The integration of this approach with a hyaluronic acid-fibrin (HA-FB) hydrogel scaffold is also studied, thus expanding the potential of NGs applications in regenerative medicine. The study shows robust antimiR encapsulation in the NGs using electroporation and the NGs ability to be internalized in MSCs and to deliver their cargo while avoiding endo-lysosomal degradation. This leads to rapid and strong knock-down of miR-221 in hMSCs in vitro, both in 2D and 3D hydrogel culture conditions (>90% and > 80% silencing efficiency, respectively). Finally, in vivo studies performed with an osteochondral defect model demonstrate the NGs ability to effectively deliver antimiR to endogenous cells. Altogether, these results prove that the NGs can operate as stand-alone system or as integrated platform in combination with scaffolds for the delivery of ASOs for a wide range of applications in drug delivery and regenerative medicine.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Biomimetics , Humans , Hydrogels , Oligonucleotides, Antisense
5.
Cell Stem Cell ; 28(4): 653-670.e11, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33561425

ABSTRACT

Bone marrow stromal cells (BMSCs) play pivotal roles in tissue maintenance and regeneration. Their origins, however, remain incompletely understood. Here we identify rare LNGFR+ cells in human fetal and regenerative bone marrow that co-express endothelial and stromal markers. This endothelial subpopulation displays transcriptional reprogramming consistent with endothelial-to-mesenchymal transition (EndoMT) and can generate multipotent stromal cells that reconstitute the bone marrow (BM) niche upon transplantation. Single-cell transcriptomics and lineage tracing in mice confirm robust and sustained contributions of EndoMT to bone precursor and hematopoietic niche pools. Interleukin-33 (IL-33) is overexpressed in subsets of EndoMT cells and drives this conversion process through ST2 receptor signaling. These data reveal generation of tissue-forming BMSCs from mouse and human endothelial cells and may be instructive for approaches to human tissue regeneration.


Subject(s)
Bone Marrow , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Cells , Endothelial Cells , Endothelium , Hematopoietic Stem Cells , Mice , Stromal Cells
6.
J Control Release ; 328: 985-999, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32860929

ABSTRACT

Osteoarthritis (OA) and intervertebral disc degeneration (IVDD) as major cause of chronic low back pain represent the most common degenerative joint pathologies and are leading causes of pain and disability in adults. Articular cartilage (AC) and intervertebral discs are cartilaginous tissues with a similar biochemical composition and pathophysiological aspects of degeneration. Although treatments directed at reversing these conditions are yet to be developed, many promising disease-modifying drug candidates are currently under investigation. Given the localized nature of these chronic diseases, drug delivery systems have the potential to enhance therapeutic outcomes by providing controlled and targeted release of bioactives, minimizing the number of injections needed and increasing drug concentration in the affected areas. This review provides a comprehensive overview of the currently most promising disease-modifying drugs as well as potential drug delivery systems for OA and IVDD therapy.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Osteoarthritis , Pharmaceutical Preparations , Adult , Drug Delivery Systems , Humans , Intervertebral Disc Degeneration/drug therapy , Osteoarthritis/drug therapy
7.
J Control Release ; 309: 220-230, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31369767

ABSTRACT

Articular cartilage is frequently injured by trauma or osteoarthritis, with limited and inadequate treatment options. We investigated a new strategy based on hydrogel-mediated delivery of a locked nucleic acid microRNA inhibitor targeting miR-221 (antimiR-221) to guide in situ cartilage repair by endogenous cells. First, we showed that transfection of antimiR-221 into human bone marrow-derived mesenchymal stromal cells (hMSCs) blocked miR-221 expression and enhanced chondrogenesis in vitro. Next, we loaded a fibrin/hyaluronan (FB/HA) hydrogel with antimiR-221 in combination or not with lipofectamine carrier. FB/HA strongly retained functional antimiR-221 over 14 days of in vitro culture, and provided a supportive environment for cell transfection, as validated by flow cytometry and qRT-PCR analysis. Seeding of hMSCs on the surface of antimiR-221 loaded FB/HA led to invasion of the hydrogel and miR-221 knockdown in situ within 7 days. Overall, the use of lipofectamine enhanced the potency of the system, with increased antimiR-221 retention and miR-221 silencing in infiltrating cells. Finally, FB/HA hydrogels were used to fill defects in osteochondral biopsies that were implanted subcutaneously in mice. FB/HA loaded with antimiR-221/lipofectamine significantly enhanced cartilage repair by endogenous cells, demonstrating the feasibility of our approach and the need to achieve highly effective in situ transfection. Our study provides new evidence on the treatment of focal cartilage injuries using controlled biomaterial-mediated delivery of antimicroRNA for in situ guided regeneration.


Subject(s)
Chondrogenesis , Drug Delivery Systems/methods , Hydrogels/chemistry , MicroRNAs/administration & dosage , Aged , Animals , Cartilage, Articular/injuries , Cartilage, Articular/physiology , Cells, Cultured , Female , Fibrin/chemistry , Humans , Hyaluronic Acid/chemistry , Mesenchymal Stem Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/therapeutic use , Middle Aged , Regeneration
8.
J Orthop Res ; 37(1): 12-22, 2019 01.
Article in English | MEDLINE | ID: mdl-30175861

ABSTRACT

Trauma and age-related cartilage disorders represent a major global cause of morbidity, resulting in chronic pain and disability in patients. A lack of effective therapies, together with a rapidly aging population, creates an impressive clinical and economic burden on healthcare systems. In this scenario, experimental therapies based on transplantation or in situ stimulation of skeletal Mesenchymal Stem/progenitor Cells (MSCs) have raised great interest for cartilage repair. Nevertheless, the challenge of guiding MSC differentiation and preventing cartilage hypertrophy and calcification still needs to be overcome. While research has mostly focused on the stimulation of cartilage anabolism using growth factors, several issues remain unresolved prompting the field to search for novel solutions. Recently, inhibition of anti-chondrogenic regulators has emerged as an intriguing opportunity. Anti-chondrogenic regulators include extracellular proteins as well as intracellular transcription factors and microRNAs that act as potent inhibitors of pro-chondrogenic signals. Suppression of these inhibitors can enhance MSC chondrogenesis and production of cartilage matrix. We here review the current knowledge concerning different types of anti-chondrogenic regulators. We aim to highlight novel therapeutic targets for cartilage repair and discuss suitable tools for suppressing their anti-chondrogenic functions. Further effort is needed to unveil the therapeutic perspectives of this approach and pave the way for effective treatment of cartilage injuries in patients. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.


Subject(s)
Chondrogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Molecular Targeted Therapy , Animals , Cartilage/injuries , Humans , Mesenchymal Stem Cell Transplantation
9.
Article in English | MEDLINE | ID: mdl-28660185

ABSTRACT

Tissue engineering (TE) approaches using biomaterials have gain important roles in the regeneration of cartilage. This paper describes the production by microfluidics of alginate-based microfibers containing both extracellular matrix (ECM)-derived biomaterials and chondrocytes. As ECM components gelatin or decellularized urinary bladder matrix (UBM) were investigated. The effectiveness of the composite microfibers has been tested to modulate the behavior and redifferentiation of dedifferentiated chondrocytes. The complete redifferentiation, at the single-cell level, of the chondrocytes, without cell aggregate formation, was observed after 14 days of cell culture. Specific chondrogenic markers and high cellular secretory activity was observed in embedded cells. Notably, no sign of collagen type 10 deposition was determined. The obtained data suggest that dedifferentiated chondrocytes regain a functional chondrocyte phenotype when embedded in appropriate 3D scaffold based on alginate plus gelatin or UBM. The proposed scaffolds are indeed valuable to form a cellular microenvironment mimicking the in vivo ECM, opening the way to their use in cartilage TE.

10.
Cell Mol Life Sci ; 74(19): 3451-3465, 2017 10.
Article in English | MEDLINE | ID: mdl-28434038

ABSTRACT

The field of cartilage repair has exponentially been growing over the past decade. Here, we discuss the possibility to achieve satisfactory regeneration of articular cartilage by means of human mesenchymal stem cells (hMSCs) depleted of anti-chondrogenic factors and implanted in the site of injury. Different types of molecules including transcription factors, transcriptional co-regulators, secreted proteins, and microRNAs have recently been identified as negative modulators of chondroprogenitor differentiation and chondrocyte function. We review the current knowledge about these molecules as potential targets for gene knockdown strategies using RNA interference (RNAi) tools that allow the specific suppression of gene function. The critical issues regarding the optimization of the gene silencing approach as well as the delivery strategies are discussed. We anticipate that further development of these techniques will lead to the generation of implantable hMSCs with enhanced potential to regenerate articular cartilage damaged by injury, disease, or aging.


Subject(s)
Cartilage, Articular/physiology , Chondrogenesis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , RNA Interference , RNAi Therapeutics/methods , Regeneration , Animals , Cartilage, Articular/injuries , Humans , Mesenchymal Stem Cell Transplantation/methods , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , RNA, Untranslated/genetics , Transcription Factors/genetics
11.
Life Sci ; 152: 82-93, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27015789

ABSTRACT

AIMS: We aimed to establish a 3D osteoblasts/osteoclasts co-culture system requiring limited amounts of human primary cells and useful as platform to 1. recapitulate an "oral bone microenvironment" in healthy or pathological condition, and 2. produce potential implantable cell constructs for regeneration of jawbone which can be negatively affected by bisphosphonates (BPs). MAIN METHODS: Osteoblasts from normal bone chips (hOBs) or from jawbone of patients taking BPs (hnOBs) were co-cultured with monocytes (hMCs) either in static (3D-C) or dynamic (3D-DyC) condition using the RCCS-4™ bioreactor for 3weeks. Cell aggregates were characterized for viability, histological features and specific osteoclastic and osteogenic markers. KEY FINDINGS: In all tested conditions hOBs supported the formation of mature osteoclasts (hOCs), without differentiating agents or exogenous scaffolds. 3D-DyC condition associated with a ground based condition (Xg) rather than modeled microgravity (µXg) produced aggregates with high level of osteogenic markers including Osteopontin (OPN), Osterix (OSX), Runx2 and appreciable bone mineral matrix. hnOBs co-cultured with hMCs in 3D-Dyc/Xg condition generated OPN and mineral matrix positive aggregates. SIGNIFICANCE: We optimized a 3D co-culture system with a limited amount of cells preserving viability and functionality of bone cellular components and generating bone-like aggregates also by using cells from jawbone necrotic tissue. The feasibility to obtain from poor-quality bone sites viable osteoblasts able to form aggregates when co-cultured with hMCs, allows to study the development of autologous implantable constructs to overcome jawbone deficiency in patients affected by MRONJ (Medication-Related Osteonecrosis of the Jaws).


Subject(s)
Jaw/cytology , Osteoblasts/physiology , Osteoclasts/physiology , Aged , Aged, 80 and over , Biomarkers/metabolism , Bone Density Conservation Agents/pharmacology , Bone and Bones/cytology , Cell Survival/drug effects , Coculture Techniques , Diphosphonates/pharmacology , Environment , Female , Humans , Male , Mandible/cytology , Middle Aged , Monocytes/drug effects , Necrosis , Osteoblasts/drug effects , Osteoclasts/drug effects , Weightlessness
12.
Stem Cells ; 34(7): 1801-11, 2016 07.
Article in English | MEDLINE | ID: mdl-26930142

ABSTRACT

There is a growing demand for the development of experimental strategies for efficient articular cartilage repair. Current tissue engineering-based regenerative strategies make use of human mesenchymal stromal cells (hMSCs). However, when implanted in a cartilage defect, control of hMSCs differentiation toward the chondrogenic lineage remains a significant challenge. We have recently demonstrated that silencing the antichondrogenic regulator microRNA-221 (miR-221) was highly effective in promoting in vitro chondrogenesis of monolayered hMSCs in the absence of the chondrogenic induction factor TGF-ß. Here we investigated the feasibility of this approach first in conventional 3D pellet culture and then in an in vivo model. In pellet cultures, we observed that miR-221 silencing was sufficient to drive hMSCs toward chondrogenic differentiation in the absence of TGF-ß. In vivo, the potential of miR-221 silenced hMSCs was investigated by first encapsulating the cells in alginate and then by filling a cartilage defect in an osteochondral biopsy. After implanting the biopsy subcutaneously in nude mice, we found that silencing of miR-221 strongly enhanced in vivo cartilage repair compared to the control conditions (untreated hMSCs or alginate-only). Notably, miR-221 silenced hMSCs generated in vivo a cartilaginous tissue with no sign of collagen type X deposition, a marker of undesired hypertrophic maturation. Altogether our data indicate that silencing miR-221 has a prochondrogenic role in vivo, opening new possibilities for the use of hMSCs in cartilage tissue engineering. Stem Cells 2016;34:1801-1811.


Subject(s)
Cartilage/pathology , Chondrogenesis , Gene Silencing , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Wound Healing , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Humans , Mice, Nude , MicroRNAs/genetics , Models, Biological , Regeneration
13.
Stem Cell Rev Rep ; 10(6): 841-55, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24923882

ABSTRACT

In this study we have inhibited the expression of two negative regulators of chondrogenesis, Slug transcription factor (TF) and the small non-coding single stranded RNA microRNA-221 (miR-221), in human mesenchymal stem cells (MSCs). Our aim was test a new approach to guide the cells toward a chondrocyte - like phenotype, without the employment of differentiating agents, in the prospect of their clinical applications for cell-based cartilage tissue engineering. We have characterized these manipulated cells by gene expression analysis at the RNA and protein levels. We demonstrated that decreased miR-221 or Slug induced an increase of chondrogenic markers, including collagen type II (Col2A1), and the positive chondrogenic TFs Sox9 and TRPS1. Slug and TRPS1 are not direct targets of miR-221 since their expression was not affected by miR-221 content. Further, we showed by gene expression and Chromatin Immunoprecipitation analyses that i. miR-221 is positively regulated by Slug in hMSCs, where Slug and miR-221 high levels hamper cell differentiation, and ii. TRPS1 contributes to maintaining low levels of miR-221, both in hMSCs committed toward chondrogenesis by Slug depletion and in chondrocytes, where the low levels of miR-221 and Slug allow a chondrogenic phenotype.Taken together, our data may be relevant both to understand yet unknown miRNA - TF regulatory loops in cartilage biology and to establish new strategies based on a siRNA approach for cartilage tissue engineering.


Subject(s)
Chondrogenesis/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Transcription Factors/metabolism , Chondrocytes/metabolism , Chondrocytes/physiology , Chondrogenesis/physiology , Humans , Mesenchymal Stem Cells/physiology , Snail Family Transcription Factors
14.
Tissue Eng Part A ; 20(19-20): 2795-805, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24712489

ABSTRACT

The use of short interfering RNA (siRNA) in combination with stem cells and biocompatible scaffolds is a promising strategy in regenerative medicine. Our experimental strategy was to explore the possibility of forcing or guiding the chondrogenic differentiation of human mesenchymal stem cells (hMSCs) by knocking down a negative regulator of chondrogenesis, Slug transcription factor (TF), thus altering cell behavior. We found that TGFß-driven chondrogenic differentiation of hMSCs cultured onto a hyaluronan-based scaffold, HYAFF(®)-11, was strengthened after cell exposure to siRNA against Slug. Slug silencing was effective in promoting the expression of chondrogenic markers, including Col2A1, aggrecan, Sox9, LEF1, and TRPS1. In addition, we confirmed that HYAFF-11 is a good scaffold candidate for hMSC use in tissue engineering applications, and showed that it is effective in sustaining TGFß3 treatment associated with a specific gene silencing. Interestingly, preliminary results from the experimental model described here suggested that, even in the absence of differentiation supplements, Slug silencing showed a pro-chondrogenic effect, highlighting both its potential use as an alternative to TGFß treatment, and the critical role of the Slug TF in determining the fate of hMSCs.


Subject(s)
Cell Differentiation , Chondrogenesis , Gene Silencing , Mesenchymal Stem Cells/metabolism , RNA, Small Interfering , Transcription Factors/metabolism , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Snail Family Transcription Factors , Transcription Factors/genetics
15.
BMC Cancer ; 12: 445, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-23031797

ABSTRACT

BACKGROUND: Breast cancer and its metastatic progression is mainly directed by epithelial to mesenchymal transition (EMT), a phenomenon supported by specific transcription factors and miRNAs. METHODS: In order to investigate a possible correlation between Slug transcription factor and miR-221, we performed Slug gene silencing in MDA-MB-231 breast cancer cells and evaluated the expression of genes involved in supporting the breast cancer phenotype, using qRT-PCR and Western blot analysis. Chromatin immunoprecipitation and wound healing assays were employed to determine a functional link between these two molecules. RESULTS: We showed that Slug silencing significantly decreased the level of miR-221 and vimentin, reactivated Estrogen Receptor α and increased E-cadherin and TRPS1 expression. We demonstrated that miR-221 is a Slug target gene, and identified a specific region of miR-221 promoter that is transcriptionally active and binds the transcription factor Slug "in vivo". In addition, we showed that in Slug-silenced cells, wich retained residual miR-221 (about 38%), cell migration was strongly inhibited. Cell migration was inhibited, but to a less degree, following complete knockdown of miR-221 expression by transfection with antagomiR-221. CONCLUSIONS: We report for the first time evidence of a correlation between Slug transcription factor and miR-221 in breast cancer cells. These studies suggest that miR-221 expression is, in part, dependent on Slug in breast cancer cells, and that Slug plays a more important role than miR-221 in cell migration and invasion.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Transcription Factors/genetics , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Chromatin Immunoprecipitation , Female , Gene Silencing , Humans , MicroRNAs/metabolism , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/metabolism , Transfection
16.
Int J Mol Med ; 28(2): 199-206, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21584488

ABSTRACT

The present study describes, for the first time, the removal of the nuclear factor of activated T cells cytoplasmic 1 (NFATc1) by a decoy approach in human primary osteoblasts (hOBs). hOBs with different NFATc1 expression levels were used. The functionality of endogenous NFAT proteins in our experimental model was analyzed by monitoring the transcriptional activity on a luciferase reporter construct driven by three copies of an NFAT response element (pNFAT-TA-luc). Cell treatment with the decoy against NFATc1 resulted in a significant increase in the expression of osteoblastic markers, including ERα and ColXV. On the contrary, the expression of Runx2, which is known to not be transcriptionally regulated by NFATc1, was not altered, indicating the specificity of the decoy effect. To our knowledge, this is the first time that transcription factor decoy has been successful in hOBs to allow the investigation of the role of NFATc1 in an experimental model that, compared to the use of cell lines, more closely resembles an in vivo model. In addition, by using chromatin immunoprecipitation we found that in vivo NFATc1 is recruited on the ColXV gene promoter. The specific role of NFATc1 in osteoblast differentiation is not well understood, however, our findings reinforce the action of NFATc1 in the transcriptional program of osteoblasts, also supporting the therapeutic potential for the proper manipulation of NFATc1-mediated events in different bone disorders. At the same time, our data add important information on the regulation of the expression of ColXV, which only recently has been proposed as an osteoblastic marker.


Subject(s)
NFATC Transcription Factors/metabolism , Osteoblasts/metabolism , Transcription Factors/metabolism , Aged , Cell Differentiation/genetics , Cells, Cultured , Collagen/genetics , Gene Expression Regulation , Gene Silencing , Humans , Jurkat Cells , Middle Aged , NFATC Transcription Factors/genetics , Oligonucleotides/genetics , Oligonucleotides/metabolism , Osteoblasts/cytology , Promoter Regions, Genetic , Response Elements/genetics , Transcription Factors/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...