dfat cells
Recently Published Documents


TOTAL DOCUMENTS

28
(FIVE YEARS 11)

H-INDEX

8
(FIVE YEARS 0)

2021 ◽  
Vol 11 (24) ◽  
pp. 11941
Author(s):  
Nobuhito Tsumano ◽  
Hirohito Kubo ◽  
Rie Imataki ◽  
Yoshitomo Honda ◽  
Yoshiya Hashimoto ◽  
...  

Mechanical and resorbable scaffolds are in high demand for stem cell-based regenerative medicine, to treat refractory bone defects in craniofacial abnormalities and injuries. Multipotent progenitor cells, such as dedifferentiated fat (DFAT) cells, are prospective sources for regenerative therapies. Herein, we aimed to demonstrate that a composite gelatin sponge (α-TCP/GS) of alfa-tricalcium phosphate (α-TCP) mixed with gelatin scaffolds (GS), with/without DFATs, induced bone regeneration in a rat calvarial defect model in vivo. α-TCP/GS was prepared by mixing α-TCP and 2% GS using vacuum-heated methods. α-TCP/GS samples with/without DFATs were transplanted into the model. After 4 weeks of implantation, the samples were subjected to micro-computed tomography (μ-CT) and histological analysis. α-TCP/GS possessed adequate mechanical strength; α-TCP did not convert to hydroxyapatite upon contact with water, as determined by X-ray diffraction. Moreover, stable α-TCP/GS was formed by electrostatic interactions, and verified based on the infrared peak shifts. μ-CT analyses showed that bone formation was higher in the α-TCP/GS+ DFAT group than in the α-TCP/GS group. Therefore, the implantation of α-TCP/GS comprising DFAT cells enhanced bone regeneration and vascularization, demonstrating the potential for healing critical-sized bone defects.


2021 ◽  
Vol 22 (22) ◽  
pp. 12392
Author(s):  
Kiyofumi Takabatake ◽  
Masakazu Matsubara ◽  
Eiki Yamachika ◽  
Yuki Fujita ◽  
Yuki Arimura ◽  
...  

Background: We investigated and compared the osteogenic potential and bone regeneration capacities of dedifferentiated fat cells (DFAT cells) and adipose-derived stem cells (ASCs). Method: We isolated DFAT cells and ASCs from GFP mice. DFAT cells were established by a new culture method using a mesh culture instead of a ceiling culture. The isolated DFAT cells and ASCs were incubated in osteogenic medium, then alizarin red staining, alkaline phosphatase (ALP) assays, and RT-PCR (for RUNX2, osteopontin, DLX5, osterix, and osteocalcin) were performed to evaluate the osteoblastic differentiation ability of both cell types in vitro. In vivo, the DFAT cells and ASCs were incubated in osteogenic medium for four weeks and seeded on collagen composite scaffolds, then implanted subcutaneously into the backs of mice. We then performed hematoxylin and eosin staining and immunostaining for GFP and osteocalcin. Results: The alizarin red-stained areas in DFAT cells showed weak calcification ability at two weeks, but high calcification ability at three weeks, similar to ASCs. The ALP levels of ASCs increased earlier than in DFAT cells and showed a significant difference (p < 0.05) at 6 and 9 days. The ALP levels of DFATs were higher than those of ASCs after 12 days. The expression levels of osteoblast marker genes (osterix and osteocalcin) of DFAT cells and ASCs were higher after osteogenic differentiation culture. Conclusion: DFAT cells are easily isolated from a small amount of adipose tissue and are readily expanded with high purity; thus, DFAT cells are applicable to many tissue-engineering strategies and cell-based therapies.


2021 ◽  
Author(s):  
Takashi Maruyama ◽  
Noboru Fukuda ◽  
Taro Matsumoto ◽  
Morito Endo ◽  
Kei Utsunomiya ◽  
...  

Abstract Background We established an adipogenic progenitor cell line derived from mature adipocytes and named these cells dedifferentiated fat (DFAT) cells, which have been shown to have characteristics very similar to those of mesenchymal stem cells (MSCs). The potential application of DFAT cells to support cell-based therapies for regenerative and immunosuppressive therapies has been suggested. The present study was designed to address beneficial ways that DFAT implantation can be used clinically as immunosuppressive therapy to treat immunological glomerulonephritis. Methods We evaluated distribution of DFAT cells after intravenous injection through the tail vein in Wistar rats. We examined effects of allogenic implantation of DFAT cells on BrdU incorporation into kidney from rats with monoclonal antibody (mAb) 1-22-3-induced glomerulonephritis. We compared effects of allogenic and autogenic implantations of DFAT cells on excretion of urinary protein, renal function, and glomerular and nephrotubular injuries in these rats, and serum levels of tumor necrosis factor-stimulated gene-6 (TSG-6), and expression of TSG-6 mRNA in kidney. Results The allogenic implantations of DFAT cells trapped in lung improved excretion of urinary protein and renal function, and significantly suppressed glomerular and nephrotubular injuries in the rats with mAb1-22-3-induced glomerulonephritis compared with the autogenic implantations. The allogenic implantation of DFAT cells increased serum levels of TSG-6 especially in mAb 1-22-3-induced glomerulonephritis and significantly increased the expression of TSG-6 mRNA in kidney compared to the autogenic implantation. Conclusion These findings suggest that allogenic implantation of DFAT cells could be clinically useful immunosuppressive therapy for immunological glomerulonephritis.


2021 ◽  
Author(s):  
Kei Utsunomiya ◽  
Takashi Maruyama ◽  
Satoshi Shimizu ◽  
Taro Matsumoto ◽  
Morito Endo ◽  
...  

Abstract We examined the effects of implantation of dedifferentiated fat (DFAT) cells on renal function, proteinuria and glomerulonephritis in SCG mice as a preclinical study of DFAT cell therapy for antineutrophil cytoplasmic antibody (ANCA) glomerulonephritis and investigated mechanisms underlying the immunosuppressive effects of the implantation of DFAT cells. After their intravenous infusion, almost all DFAT cells were trapped in the lung and not delivered into the kidney. Implantation of DFAT cells in SCG mice suppressed glomerular crescent formation, decreased urinary protein excretions, and increased expression of tumor necrosis factor-stimulated gene-6 (TSG-6) mRNA, protein and immunostaining in kidney from these mice. Implantation of DFAT cells increased the expression of microRNA 23b-3p in plasma, kidney and lung in SCG mice and decreased the expression of CD44 mRNA and increased the expression of prostaglandin E2 and interleukin-10 mRNAs in kidney from these mice. Implantation of DFAT cells increased expression of TSG-6 protein and decreased expression of tumor necrosis factor-a protein in kidney from SCG mice. Further, implantation of DFAT cells increased the expression of C-C motif chemokine ligand 17 protein, a chemokine for M2 macrophages, and decreased the expression of MCP-1 protein, a chemokine for M1 macrophages, in kidney from SCG mice. Survival rates were higher in SCG mice with implantation of DFAT cells than in SCG mice without implantation. These results indicate that implantation of DFAT cells suppressed renal injury including glomerular crescent formation in kidney from SCG mice while increasing the expression of TSG-6 without delivery of DFAT cells directly into kidney. Mechanisms underlying the effects of improvement of ANCA glomerulonephritis are associated with immunosuppressive effects by TSG-6 and the transition of M1 to M2 macrophages. These findings suggest that implantation of DFAT cells may become a cell therapy for ANCA glomerulonephritis.


Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1475
Author(s):  
Anna Figiel-Dabrowska ◽  
Klaudia Radoszkiewicz ◽  
Paulina Rybkowska ◽  
Natalia Ewa Krzesniak ◽  
Dorota Sulejczak ◽  
...  

Currently, the number of stem-cell based experimental therapies in neurological injuries and neurodegenerative disorders has been massively increasing. Despite the fact that we still have not obtained strong evidence of mesenchymal stem/stromal cells’ neurogenic effectiveness in vivo, research may need to focus on more appropriate sources that result in more therapeutically promising cell populations. In this study, we used dedifferentiated fat cells (DFAT) that are proven to demonstrate more pluripotent abilities in comparison with standard adipose stromal cells (ASCs). We used the ceiling culture method to establish DFAT cells and to optimize culture conditions with the use of a physioxic environment (5% O2). We also performed neural differentiation tests and assessed the neurogenic and neuroprotective capability of both DFAT cells and ASCs. Our results show that DFAT cells may have a better ability to differentiate into oligodendrocytes, astrocytes, and neuron-like cells, both in culture supplemented with N21 and in co-culture with oxygen–glucose-deprived (OGD) hippocampal organotypic slice culture (OHC) in comparison with ASCs. Results also show that DFAT cells have a different secretory profile than ASCs after contact with injured tissue. In conclusion, DFAT cells constitute a distinct subpopulation and may be an alternative source in cell therapy for the treatment of nervous system disorders.


Antioxidants ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 905
Author(s):  
Gitit Kra ◽  
Jayasimha Rayalu Daddam ◽  
Hadar Gabay ◽  
Sara Yosefi ◽  
Maya Zachut

Heat stress (HS) induces oxidative stress by increasing reactive oxygen species (ROS), and the polyphenol resveratrol (RSV) has been shown to have antioxidant properties by reducing ROS. Hence, we aimed to examine the effects of RSV, HS and their interaction on bovine adipocytes. We generated bovine dedifferentiated adipocyte-derived progeny (DFAT) cells from subcutaneous adipose tissue and examined the effects of RSV (100 µM), heat conditions: isothermal (ISO-37 °C), short heat (SH-41.2 °C for 1 h) and long HS (LH-41.2 °C for 16 h), and their interaction on gene expression in DFAT-cells. In medium of DFAT-cells treated with RSV, malondialdehyde levels were reduced and oxygen-radical absorbance-capacity levels were increased compared to control. Treating DFAT-cells with RSV increased the relative mRNA expression of stress-induced-phosphoprotein-1 (STIP1) and the expression of hormone-sensitive-lipase (LIPE) and perilipin-1 (PLIN1), whereas it reduced the expressions of fatty-acid-synthase (FASN) and of pro-inflammatory chemotactic-C-C-motif-chemokine-ligand-2 (CCL2) also under HS. Moreover, reduced protein abundance of FASN was found in RSV-treated DFAT-cells compared to controls. Molecular docking of RSV with FASN confirmed its possible binding to FASN active site. This work demonstrates that RSV has an antioxidant effect on bovine DFAT cells and may induce adipose lipolysis and reduce lipogenesis also under in vitro HS conditions.


Materials ◽  
2020 ◽  
Vol 13 (22) ◽  
pp. 5097
Author(s):  
Kosuke Nakano ◽  
Hirohito Kubo ◽  
Masahiro Nakajima ◽  
Yoshitomo Honda ◽  
Yoshiya Hashimoto

Bone regeneration using mesenchymal stem cells has several limitations. We investigated adipose-derived dedifferentiated fat (DFAT) cells as an alternative, and evaluated their cell proliferation rate, osteoblast differentiation, and bone regeneration ability in combination with activated platelet-rich plasma (aPRP). Rat DFATs and aPRP were isolated using ceiling culture and centrifugation, respectively. The cell proliferation rate was measured, and the cells were cultured in an osteoblast differentiation medium under varying concentrations of aPRP for 21 days and stained with Alizarin red. Gene expression was evaluated using real time polymerase chain reaction. Critical defects were implanted with DFAT seeded gelatin sponges under aPRP, and four weeks later, the bone regeneration ability was evaluated using micro-computed tomography and hematoxylin-eosin staining. The cell proliferation rate was significantly increased by the addition of aPRP. Alizarin red staining was positive 21 days after the start of induction, with significantly higher Runt-related transcription factor 2 (Runx2) and osteocalcin (OCN) expression levels than those in the controls. A 9 mm critical defect was largely closed (60.6%) after four weeks of gelatin sponge implantation with DFAT and aPRP. Therefore, materials combining DFAT cells and aPRP may be an effective approach for bone regeneration. Further research is needed to explore the long-term effects of these materials.


Human Cell ◽  
2020 ◽  
Vol 33 (4) ◽  
pp. 974-989
Author(s):  
Kallapat Tansriratanawong ◽  
Isao Tabei ◽  
Hiroshi Ishikawa ◽  
Akihiro Ohyama ◽  
Junko Toyomura ◽  
...  

Abstract Adipose-derived stem cells (ASCs) and dedifferentiated fat (DFAT) cells are alternative cell sources in tissue engineering and regeneration because they are easily obtained and exhibit multilineage differentiation. However, aging may attenuate their regenerative potential and metabolic functions. Reports characterizing DFAT cells derived from aging donors are rare, and comparisons of DNA methylation profiles between aging ASCs and DFAT cells are poorly understood. Therefore, this study aimed to characterize DFAT cells relative to ASCs derived from aging subjects and compare the DNA methylation profiles of four adipogenic genes in these cells. ASCs and DFAT cells from aging donors exhibited characteristics similar to those of stem cells, including colony formation, proliferation, and multilineage differentiation abilities. However, compared with ASCs, DFAT cells exhibited increased proliferation, smooth muscle actin alpha (SMA-α) expression and decreased cellular senescence. DNA methylation profiling of ASCs and DFAT cells by combined bisulfite restriction analysis (COBRA) demonstrated hypermethylation patterns in three potent adipogenic genes—peroxisome proliferator-activated receptor gamma 2 (PPARγ2), fatty acid-binding protein 4 (FABP4), and lipoprotein lipase (LPL)—but hypomethylation of CCAAT/enhancer binding protein alpha (C/EBPα) in the aging group. Statistically significant differences were observed between the aging group and the young group. Epigenetic regulation maintains the stability of ASCs and DFAT cells in an age-dependent manner. Our findings suggested that although the DNA methylation patterns of three adipogenic genes correlated with hypermethylation and aging, ASCs and DFAT cells exhibited cellular stability and several stem cell characteristics, offering further opportunities for personalized regeneration and energy maintenance by adipogenesis during aging.


2020 ◽  
Vol 36 (7) ◽  
pp. 799-807
Author(s):  
Shigeki Ishioka ◽  
Takashi Hosokawa ◽  
Taro Ikeda ◽  
Noriyoshi Konuma ◽  
Hide Kaneda ◽  
...  

Abstract Purpose Our previous studies demonstrated that mature adipocyte-derived dedifferentiated fat (DFAT) cells possess similar multipotency as mesenchymal stem cells. Here, we examined the immunoregulatory potential of DFAT cells in vitro and the therapeutic effect of DFAT cell transplantation in a mouse inflammatory bowel disease (IBD) model. Methods The effect of DFAT cell co-culture on T cell proliferation and expression of immunosuppression-related genes in DFAT cells were evaluated. To create IBD, CD4+CD45RBhigh T cells were intraperitoneally injected into SCID mice. One week later, DFAT cells (1 × 105, DFAT group) or saline (Control group) were intraperitoneally injected. Subsequently bodyweight was measured every week and IBD clinical and histological scores were evaluated at 5 weeks after T cell administration. Results The T cell proliferation was inhibited by co-cultured DFAT cells in a cell density-dependent manner. Gene expression of TRAIL, IDO1, and NOS2 in DFAT cells was upregulated by TNFα stimulation. DFAT group improved IBD-associated weight loss, IBD clinical and histological scores compared to Control group. Conclusion DFAT cells possess immunoregulatory potential and the cell transplantation promoted recovery from colon damage and improved clinical symptoms in the IBD model. DFAT cells could play an important role in the treatment of IBD.


2020 ◽  
Author(s):  
Wei Lu ◽  
Yehua Yu ◽  
Shaoxin Yang ◽  
Yuanmei Zhai ◽  
Chong Zhao ◽  
...  

Abstract Background Dedifferentiated fat (DFAT) cells have been studied as a potential tool for various aspects of tissue engineering. However, the understanding of DFAT cells is mainly derived from studies of inguinal subcutaneous or visceral white adipose tissues rather than human bone marrow (BM). Here, we focused on DFAT cells dedifferentiated from bone marrow adipocytes. Methods A plate ceiling culture method and light microscope were used to observe the morphological changes in bone marrow adipocytes. RT-qPCR analysis, immunofluorescence and immunocytochemistry were used to compare gene and protein expression before and after the adipogenic and osteogenic differentiation of BM-DFAT cells and mesenchymal stem cells from bone marrow (BM-MSCs). CCK8 assays and annexin V-PI staining were used to assess the effect of BM-DFAT cells on the proliferation and chemoresistance of leukaemic cells. Results We found that bone marrow adipocytes can dedifferentiate into fibroblast-like multipotent cells, and BM-DFAT cells exhibited similar a morphology, immunophenotype, multipotent gene expression, proliferation ability and osteogenic differentiation in comparison with BM-MSCs. Interestingly, BM-DFAT cells possessed a higher potential for adipogenic differentiation than BM-MSCs, since they expressed a lower level of Pref-1, an inhibitor of adipogenesis. Meanwhile, our results revealed the significantly enhanced proliferation of leukaemic cell lines in the BM-DFAT cell group compared with that in the BM-MSC group. However, the effects of BM-DFAT cells were not as obvious as those of BM-MSCs on the chemoresistance of leukaemic cells. Conclusions This is the first study to illustrate the properties of BM-DFAT cells. We determined that BM-DFAT cells may be a useful cell source to study the role of bone marrow adipocytes and might be utilized for clinical applications in regenerative medicine.


Sign in / Sign up

Export Citation Format

Share Document