1
|
Xue M, Liao Y, Jiang W. Insights into the molecular changes of adipocyte dedifferentiation and its future research opportunities. J Lipid Res 2024; 65:100644. [PMID: 39303983 PMCID: PMC11550672 DOI: 10.1016/j.jlr.2024.100644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/23/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024] Open
Abstract
Recent studies have challenged the traditional belief that mature fat cells are irreversibly differentiated and revealed they can dedifferentiate into fibroblast-like cells known as dedifferentiated fat (DFAT) cells. Resembling pluripotent stem cells, DFAT cells hold great potential as a cell source for stem cell therapy. However, there is limited understanding of the specific changes that occur following adipocyte dedifferentiation and the detailed regulation of this process. This review explores the epigenetic, genetic, and phenotypic alterations associated with DFAT cell dedifferentiation, identifies potential targets for clinical regulation and discusses the current applications and challenges in the field of DFAT cell research.
Collapse
Affiliation(s)
- Mingheng Xue
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Wenqing Jiang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Karanfil AS, Louis F, Sowa Y, Matsusaki M. Cationic polymer effect on brown adipogenic induction of dedifferentiated fat cells. Mater Today Bio 2024; 27:101157. [PMID: 39113911 PMCID: PMC11304885 DOI: 10.1016/j.mtbio.2024.101157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Obesity and its associated comorbidities place a substantial burden on public health. Given the considerable potential of brown adipose tissue in addressing metabolic disorders that contribute to dysregulation of the body's energy balance, this area is an intriguing avenue for research. This study aimed to assess the impact of various polymers, including collagen type I, fibronectin, laminin, gelatin, gellan gum, and poly-l-lysine (PLL), on the in vitro brown adipogenic differentiation of dedifferentiated fat cells within a fibrin gel matrix. The findings, obtained through RT-qPCR, immunofluorescent imaging, ELISA assay, and mitochondria assessment, revealed that PLL exhibited a significant browning-inducing effect. Compared to fibrin-only brown-like drops after two weeks of incubation in brown adipogenic medium, PLL showed 6 (±3) times higher UCP1 gene expression, 5 (±2) times higher UCP1 concentration by ELISA assay, and 2 (±1) times higher mitochondrial content. This effect can be attributed to PLL's electrostatic properties, which potentially facilitate the cellular uptake of crucial brown adipogenic inducers such as the thyroid hormone, triiodothyronine (T3), and insulin from the induction medium.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Osaka University, Japan
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Osaka University, Japan
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Osaka University, Japan
| |
Collapse
|
3
|
Lim NK, Jeon HB, Kim S. The transdifferentiation of human dedifferentiated fat cells into fibroblasts: An in vitro experimental pilot study. Medicine (Baltimore) 2024; 103:e37595. [PMID: 38552064 PMCID: PMC10977558 DOI: 10.1097/md.0000000000037595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/22/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Skin grafting is a common method of treating damaged skin; however, surgical complications may arise in patients with poor health. Currently, no effective conservative treatment is available for extensive skin loss. Mature adipocytes, which constitute a substantial portion of adipose tissue, have recently emerged as a potential source of stemness. When de-lipidated, these cells exhibit fibroblast-like characteristics and the ability to redifferentiate, offering homogeneity and research utility as "dedifferentiated fat cells." METHODS AND RESULTS We conducted an in vitro study to induce fibroblast-like traits in the adipose tissue by transdifferentiating mature adipocytes for skin regeneration. Human subcutaneous fat tissues were isolated and purified from mature adipocytes that underwent a transformation process over 14 days of cultivation. Microscopic analysis revealed lipid degradation over time, ultimately transforming cells into fibroblast-like forms. Flow cytometry was used to verify their characteristics, highlighting markers such as CD90 and CD105 (mesenchymal stem cell markers) and CD56 and CD106 (for detecting fibroblast characteristics). Administering dedifferentiated fat cells with transforming growth factor-β at the identified optimal differentiation concentration of 5 ng/mL for a span of 14 days led to heightened expression of alpha smooth muscle actin and fibronectin, as evidenced by RNA and protein analysis. Meanwhile, functional validation through cell sorting demonstrated limited fibroblast marker expression in both treated and untreated cells after transdifferentiation by transforming growth factor-β. CONCLUSION Although challenges remain in achieving more effective transformation and definitive fibroblast differentiation, our trial could pave the way for a novel skin regeneration treatment strategy.
Collapse
Affiliation(s)
- Nam Kyu Lim
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan, Chungcheongnamdo, Republic of Korea
- Dankook Physician Scientist Research Center (DPSRC), Dankook University Hospital, Cheonan, Chungcheongnamdo, Republic of Korea
| | - Hong Bae Jeon
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan, Chungcheongnamdo, Republic of Korea
- Dankook Physician Scientist Research Center (DPSRC), Dankook University Hospital, Cheonan, Chungcheongnamdo, Republic of Korea
| | - Sungyeon Kim
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan, Chungcheongnamdo, Republic of Korea
| |
Collapse
|
4
|
Karanfil AS, Louis F, Sowa Y, Matsusaki M. ECM proteins and cationic polymers coating promote dedifferentiation of patient-derived mature adipocytes to stem cells. Biomater Sci 2023; 11:7623-7638. [PMID: 37830400 DOI: 10.1039/d3bm00934c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Reprogramming of mature adipocytes is an attractive research area due to the plasticity of these cells. Mature adipocytes can be reprogrammed in vitro, transforming them into dedifferentiated fat cells (DFATs), which are considered a new type of stem cell, and thereby have a high potential for use in tissue engineering and regenerative medicine. However, there are still no reports or findings on in vitro controlling the dedifferentiation. Although ceiling culture performed in related studies is a relatively simple method, its yield is low and does not allow manipulation of mature adipocytes to increase or decrease the dedifferentiation. In this study, to understand the role of physicochemical surface effects on the dedifferentiation of patient-derived mature adipocytes, the surfaces of cell culture flasks were coated with extracellular matrix, basement membrane proteins, and cationic/anionic polymers. Extracellular matrix such as fibronectin and collagen type I, and basement membrane proteins such as collagen type IV and laminin strongly promoted dedifferentiation of mature adipocytes, with laminin showing the highest effect with a DFAT ratio of 2.98 (±0.84). Interestingly, cationic polymers also showed a high dedifferentiation effect, but anionic polymers did not, and poly(diallyl dimethylammonium chloride) showed the highest DFAT ratio of 2.27 (±2.8) among the cationic polymers. Protein assay results revealed that serum proteins were strongly adsorbed on the surfaces of the cationic polymer coating, including inducing high mature adipocyte adhesion. This study demonstrates for the first time the possibility of regulating the transformation of mature adipocytes to DFAT stem cells by controlling the physicochemical properties of the surface of conventional cell culture flasks.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
5
|
Zhang X, Zhang Y, Wang L, Wu G, Pan C. Three novel simple sequence repeats (SSRs) identified by MALDI-TOF-MS method were associated with backfat in pig. Anim Biotechnol 2023; 34:1014-1021. [PMID: 35048796 DOI: 10.1080/10495398.2021.2009845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Backfat trait is an important economic trait and highly heritable, but difficult to evaluate. Thus, it is of great significance to explore optimal backfat thickness of pigs by using marker-assisted selection (MAS) to speed up its breeding process and improve economic efficiency. This study aimed to investigate the relationship between genetic variations (e.g., SSRs) and backfat of Qinghai Bamei pigs using MALDI-TOF Mass Spectrometry (MALDI-TOF-MS). Herein, five alternative SSR loci (namely V1, V2, V3, V4 and V5) were selected for subsequent detection. The results suggested that 3 (141-, 143- and 145-), 3 (128-, 130- and 132-), 2 (160- and 162-), 2 (136- and 139-) and 3 (170-, 184- and 192-) alleles of V1, V2, V3, V4 and V5 were found, respectively. Subsequent analysis showed that there was linkage equilibrium among five SSRs and Hap19 (13.1%) (141-/132-/160-/139-/192-) had the highest haplotype frequency. Among these five SSR loci, V1, V2 and V3 loci were significantly associated to the backfat of Qinghai Bamei sows. These findings enriched the study of SSRs in Qinghai Bamei pigs, and (AC)n (Chr15:85485851-85485995), (AC)n (Chr10:52724583-52724713) and (TG)n (Chr4:90732644-90732802) could be utilized as the candidate locus for MAS in pig industry.HIGHLIGHTSFive novel SSR loci was identified in pigs through MALDI-TOF MS.V1, V2 and V3 loci was were significantly associated to the backfat of pigs.
Collapse
Affiliation(s)
- Xuelian Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yanghai Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
- Meat Science and Muscle Biology Laboratory, Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Guofang Wu
- College of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Chuanying Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
6
|
Deng W, Jo JI, Morikuni H, Sasayama S, Hashimoto Y, Matsumoto N, Honda Y. Senescence-associated secretory phenotypes in rat-derived dedifferentiated fat cells with replicative senescence. Dent Mater J 2023. [PMID: 36775334 DOI: 10.4012/dmj.2022-242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Senescence-associated secretory phenotype (SASPs) secreted from senescent cells often cause the deleterious damages to the surrounding tissues. Although dedifferentiated fat (DFAT) cells prepared are considered a promising cell source for regenerative therapies, SASPs from DFAT cells undergoing long-term cell culture, which latently induce replicative senescence, have barely been explored. The present study was designed to investigate senescent behaviors in rat-derived DFAT cells at high passage numbers and to analyze the possible types of SASPs. Our data show that DFAT cells undergo senescence during replicative passaging, as determined by multiple senescent hallmarks including morphological changes in cell shape and nucleus. Moreover, RT2 PCR array analysis indicated that senescent DFAT cells expressed higher levels of 16 inflammatory cytokines (Ccl11, Ccl12, Ccl21, Ccl5, Csf2, Cxcl1, Cxcl12, Ifna2, IL11, IL12a, IL13, IL1a, IL1rn, IL6, Mif, and Tnf) associated with SASPs than non-senescent cells. This study implicates that rat DFAT cells undergo cellular senescence after long-term cell culture; cautious consideration should be paid to treat SASP secretion when senescent DFAT cells are used in regenerative medicine.
Collapse
Affiliation(s)
- Wenqi Deng
- Department of Orthodontics, Osaka Dental University
| | | | | | | | | | | | | |
Collapse
|
7
|
Bollmann A, Sons HC, Schiefer JL, Fuchs PC, Windolf J, Suschek CV. Comparative Study of the Osteogenic Differentiation Potential of Adipose Tissue-Derived Stromal Cells and Dedifferentiated Adipose Cells of the Same Tissue Origin under Pro and Antioxidant Conditions. Biomedicines 2022; 10:biomedicines10123071. [PMID: 36551827 PMCID: PMC9776284 DOI: 10.3390/biomedicines10123071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Adipose tissue-derived stromal cells (ASCs) play an important role in various therapeutic approaches to bone regeneration. However, such applications become challenging when the obtained cells show a functional disorder, e.g., an impaired osteogenic differentiation potential (ODP). In addition to ASCs, human adipose tissue is also a source for another cell type with therapeutic potential, the dedifferentiated fat cells (DFATs), which can be obtained from mature adipocytes. Here, we for the first time compared the ODPs of each donors ASC and DFAT obtained from the same adipose tissue sample as well as the role of oxidative stress or antioxidative catalase on their osteogenic outcome. Osteogenic potential of ASC and DFAT from nine human donors were compared in vitro. Flow cytometry, staining for calcium accumulation with alizarin red, alkaline phosphatase assay and Western blots were used over an osteogenic induction period of up to 14 days. H2O2 was used to induce oxidative stress and catalase was used as an antioxidative measure. We have found that ASC and DFAT cultures' ODPs are nearly identical. If ASCs from an adipose tissue sample showed good or bad ODP, so did the corresponding DFAT cultures. The inter-individual variability of the donor ODPs was immense with a maximum factor of about 20 and correlated neither with the age nor the sex of the donors of the adipose tissue. Oxidative stress in the form of exogenously added H2O2 led to a significant ODP decrease in both cell types, with this ODP decrease being significantly lower in DFAT cultures than in the corresponding ASC cultures. Regardless of the individual cell culture-specific ODP, however, exogenously applied catalase led to an approx. 2.5-fold increase in osteogenesis in the ASC and DFAT cultures. Catalase appears to be a potent pro-osteogenic factor, at least in vitro. A new finding that points to innovative strategies and therapeutic approaches in bone regeneration. Furthermore, our results show that DFATs behave similarly to ASCs of the same adipose tissue sample with respect to ODPs and could therefore be a very attractive and readily available source of multipotent stem cells in bone regenerative therapies.
Collapse
Affiliation(s)
- Anne Bollmann
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Hans Christian Sons
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Jennifer Lynn Schiefer
- Department of Plastic Surgery, Hand Surgery, Burn Center, Merheim Hospital Cologne, University of Witten/Herdecke, Ostmerheimer Straße 200, 51109 Köln, Germany
| | - Paul C. Fuchs
- Department of Plastic Surgery, Hand Surgery, Burn Center, Merheim Hospital Cologne, University of Witten/Herdecke, Ostmerheimer Straße 200, 51109 Köln, Germany
| | - Joachim Windolf
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Christoph Viktor Suschek
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
- Correspondence:
| |
Collapse
|
8
|
Tambrchi P, Mahdavi AH, DaliriJoupari M, Soltani L. Polycaprolactone-co-polylactic acid nanofiber scaffold in combination with 5-azacytidine and transforming growth factor-β to induce cardiomyocyte differentiation of adipose-derived mesenchymal stem cells. Cell Biochem Funct 2022; 40:668-682. [PMID: 35924670 DOI: 10.1002/cbf.3728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/11/2022]
Abstract
Adipose-derived mesenchymal stem cells (Ad-MSCs) are promising candidates for cardiac repair/regeneration. The application of copolymer nanoscaffolds has received great attention in tissue engineering to support differentiation and functional tissue organization toward effective tissue regeneration. The objective of the current study was to develop functional and bioactive scaffolds by combining polycaprolactone (PCL) and polylactic acid (PLA) for cardiomyocyte differentiation of human Ad-MSC (hAd-MSCs) in the absence or presence of 5-azacytidine and transforming growth factor-β (TGF-β). To that end, the human MSCs were extracted from human adipose tissue (AD). The cardiomyocyte differentiation potency of hAd-MSCs was evaluated on the novel synthetic PCL/PLA nanofiber scaffolds prepared in the absence and presence of 5-azacytidine and TGF-β supplements. A PCL/PLA nanofibrous scaffold was fabricated using the electrospinning method and its nanotopography and porous structure were characterized using scanning electron microscopy. In addition, the attachment of hAd-MSCs on the PCL/PLA scaffolds was semiquantitatively investigated. Compared with other treatments, the PCL/PLA nanofibrous scaffold supplemented with both 5-azacytidine and TGF-β was observed to differentiate hAd-MSCs into cardiomyocytes at Day 21 as evidenced by real-time PCR for cardiac-specific genes including cardiac troponin I (cTnI), GATA4, MYH7, and NKX2.5. In addition, flow cytometric analysis of cTnI-positive cells demonstrated that the cardiomyocyte differentiation of hAd-MSCs was more efficient on the PCL/PLA nanofibrous scaffold supplemented with both 5-azacytidine and TGF-β than it was in the other treatment groups. Generally speaking, the results show that PCL/PLA nanofibrous scaffolds may be applied as a platform for efficient differentiation of hAd-MSCs into functional cardiomyocytes.
Collapse
Affiliation(s)
- Parastoo Tambrchi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amir Hossein Mahdavi
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Morteza DaliriJoupari
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Leila Soltani
- Department of Animal Sciences, College of Agriculture and Natural Resources, Razi University, Kermanshah, Iran
| |
Collapse
|
9
|
Huang G, Xia B, Dai Z, Yang R, Chen R, Yang H. Comparative study of DFAT cell and ADSC sheets for periodontal tissue regeneration:
in vivo
and
in vitro
evidence. J Clin Periodontol 2022; 49:1289-1303. [PMID: 35851962 DOI: 10.1111/jcpe.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/29/2022] [Accepted: 06/30/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Guobin Huang
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Bin Xia
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Oral and Maxillofacial Surgery The Affiliated Stomatological Hospital of Kunming Medical University Kunming Yunnan P.R. China
| | - Zichao Dai
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Rongqiang Yang
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Rui Chen
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology Kunming Medical University Kunming Yunnan PR China
- Department of Dental Research The Affiliated Stomatology Hospital of Kunming Medical University Kunming Yunnan PR China
| |
Collapse
|
10
|
IGF-I and Hyaluronic Acid Mitigate the Negative Effect of Irradiation on Human Skin Keratinocytes. Cancers (Basel) 2022; 14:cancers14030588. [PMID: 35158856 PMCID: PMC8833477 DOI: 10.3390/cancers14030588] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Patients undergoing radiation therapy for the treatment of various types of cancer often experience side effects such as radiation dermatitis. A gold standard treatment is still lacking. The objective of the present study was to find novel therapeutic strategies for the regeneration and repair of damaged skin areas after irradiation. An in vitro 2D and 3D primary keratinocyte model was used to test the effect of insulin-like growth factor I (IGF-I), keratinocyte growth factor (KGF), platelet lysate (PL), hyaluronic acid (HA), and adipose-derived stem cell (ADSC) conditioned medium on the functional abilities (viability, migration) and the gene expression of irradiated keratinocytes. Hyaluronic acid and IGF-I effectively reduced the irradiation damage of primary keratinocytes by stimulating viability and migration and reducing cell apoptosis and necrosis. These findings indicate that the negative effects of irradiation on keratinocytes located in the patient’s skin can be counterbalanced with HA and IGF-I treatment. Abstract Ionizing radiation has become an integral part of modern cancer therapy regimens. Various side effects, such as radiation dermatitis, affect patients in acute and chronic forms and decrease therapy compliance significantly. In this study, primary keratinocytes were irradiated in a 2-dimensional (2D) culture as well as on a 3-dimensional (3D) collagen-elastin matrix with doses of 2 and 5 Gy. The effect of different concentrations of IGF-I, KGF, platelet lysate (PL), high and low molecular weight hyaluronic acid (H-HA, L-HA), and adipose-derived stem cell (ADSC) conditioned medium was analyzed in respect to cell viability (WST-8), wound closure (migration), and the gene expression (quantitative real-time PCR) of 2D cultures. The 3D culture was evaluated by WST-8. A mixture of H-HA and L-HA, as well as IGF-I, could significantly stimulate the keratinocyte viability and migration which were severely reduced by irradiation. The MKI67and IL6 gene expression of irradiated keratinocytes was significantly higher after H-HA/L-HA treatment. The stimulating effects of H-HA/L-HA and IGF-I were able to be confirmed in 3D culture. A positive influence on cell viability, migration, and gene expression was achieved after the treatment with H-L-HA and IGF-I. These results open the possibility of a novel therapeutic method for both the prevention and the treatment of radiation dermatitis.
Collapse
|
11
|
Nie F, Bi H, Zhang C, Ding P. Differentiation potential and mRNA profiles of human dedifferentiated adipose cells and adipose‑derived stem cells from young donors. Mol Med Rep 2020; 23:47. [PMID: 33200799 PMCID: PMC7705993 DOI: 10.3892/mmr.2020.11685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Dedifferentiated adipose cells (DAs) and adipose-derived stem cells (ADSCs) are two of the primary types of stem cells derived from adipose tissue, which have been reported to possess similar characteristics, but also exhibit unique phenotypic and functional advantages. However, several reports have described inconsistent results regarding their differences in multilineage differentiation function. Moreover, to the best of our knowledge, there are no studies assessing their myogenic ability, or the differences in the transcriptome between the two cell types derived from lipoaspirates via tumescent liposuction from the same donors. The aim of the present study was to compare the properties and expression profiles of these cell types. Subcutaneous adipose tissue of three female patients (aged 23–30 years) with a physiological BMI (19.1–23.9 kg/m2) were obtained during tumescent liposuction of the abdomen or the thigh. The stromal vascular fraction and mature adipocytes were obtained via collagenase digestion, and ADSCs and DAs were cultured successively. To determine the differences between DAs and ADSCs after 6–7 passages, cell proliferation assays, phenotypic assessment, differentiation assays and high-throughput RNA sequencing (seq) were used. Similar cell morphologies, proliferation dynamics, surface markers and transcriptome expression profiles were observed between the DAs and ADSCs. Whilst there were notable individual differences in the osteogenic, lipogenic, chondrogenic and myogenic abilities of the DAs and ADSCs, it was difficult to determine their differentiation potential based only on the cell source. Interestingly, the myogenic ability was relatively stronger in cells with relatively weaker lipogenic ability. Only 186 differentially expressed genes between the two groups were identified using RNAseq. Several of these genes were involved in biological functions such as transcription regulation, protein translation regulation, cytokine interactions and energy metabolism regulation. The results of the present study suggested a similar functional potential of DAs and ADSCs from young donors undergoing tumescent liposuction operation in regeneration areas and the balance of the differentiative ability of the same cell populations. These data may provide a foundation for further clinical administration of stem cells derived from adipose tissues in therapy.
Collapse
Affiliation(s)
- Fangfei Nie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Chen Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Pengbing Ding
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, P.R. China
| |
Collapse
|
12
|
Tang H, Chu Y, Huang Z, Cai J, Wang Z. The metastatic phenotype shift toward myofibroblast of adipose-derived mesenchymal stem cells promotes ovarian cancer progression. Carcinogenesis 2020; 41:182-193. [PMID: 31046126 DOI: 10.1093/carcin/bgz083] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 04/06/2019] [Accepted: 04/30/2019] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer metastasizes to organs in the abdominal cavity, such as the omentum that is a rich source of adipose-derived mesenchymal stem cells (ADSCs). In present, ADSCs have received more and more attention for their roles in the development of cancer. In this study, we examined α-smooth muscle actin (α-SMA) expression and carcinoma-associated fibroblast (CAF)-like differentiation capabilities in ADSCs from omentum of different patients. The effects of ADSCs on the proliferation and invasion of epithelial ovarian cancer cells (EOCCs) were also assessed in vitro and in vivo. Our results showed that ADSCs from omentum of ovarian cancer patients, no matter whether metastasis or not, expressed higher levels of α-SMA than ADSCs from patients with benign gynecologic disease. Using direct and indirect co-culture system, we found that EOCCs induced ADSCs to express CAF markers, including α-SMA and fibroblast activation protein, via the transforming growth factor beta 1 (TGF-β1) signaling pathway. Moreover, co-cultured ADSCs exhibited functional properties similar to those of CAFs, including the ability to promote EOCCs proliferation, progression and metastasis both in vitro and in vivo. Furthermore, blocking the TGF-β1 pathway can counteract the CAF-like differentiation and tumor promotion effect of ADSCs. Our results suggest that ADSCs are a source of CAFs and that they participate in the interaction between EOCCs and the omental microenvironment. EOCCs could induce ADSCs in the omentum to differentiate before ovarian cancer metastasis, which participate in the formation of omental metastatic niches and promote the proliferation and invasion of ovarian cancer.
Collapse
Affiliation(s)
- Huijuan Tang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zaiju Huang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Gene expression profiles of early chondrogenic markers in dedifferentiated fat cells stimulated by bone morphogenetic protein 4 under monolayer and spheroid culture conditions in vitro. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.odw.2016.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Sparks R, Lui A, Bader D, Patel R, Murr M, Guida W, Fratti R, Patel NA. A specific small-molecule inhibitor of protein kinase CδI activity improves metabolic dysfunction in human adipocytes from obese individuals. J Biol Chem 2019; 294:14896-14910. [PMID: 31413114 DOI: 10.1074/jbc.ra119.008777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/11/2019] [Indexed: 12/17/2022] Open
Abstract
The metabolic consequences and sequelae of obesity promote life-threatening morbidities. PKCδI is an important elicitor of inflammation and apoptosis in adipocytes. Here we report increased PKCδI activation via release of its catalytic domain concurrent with increased expression of proinflammatory cytokines in adipocytes from obese individuals. Using a screening strategy of dual recognition of PKCδI isozymes and a caspase-3 binding site on the PKCδI hinge domain with Schrödinger software and molecular dynamics simulations, we identified NP627, an organic small-molecule inhibitor of PKCδI. Characterization of NP627 by surface plasmon resonance (SPR) revealed that PKCδI and NP627 interact with each other with high affinity and specificity, SPR kinetics revealed that NP627 disrupts caspase-3 binding to PKCδI, and in vitro kinase assays demonstrated that NP627 specifically inhibits PKCδI activity. The SPR results also indicated that NP627 affects macromolecular interactions between protein surfaces. Of note, release of the PKCδI catalytic fragment was sufficient to induce apoptosis and inflammation in adipocytes. NP627 treatment of adipocytes from obese individuals significantly inhibited PKCδI catalytic fragment release, decreased inflammation and apoptosis, and significantly improved mitochondrial metabolism. These results indicate that PKCδI is a robust candidate for targeted interventions to manage obesity-associated chronic inflammatory diseases. We propose that NP627 may also be used in other biological systems to better understand the impact of caspase-3-mediated activation of kinase activity.
Collapse
Affiliation(s)
- Robert Sparks
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Illinois 61801
| | - Ashley Lui
- Department of Molecular Medicine, University of South Florida, Tampa, Florida 33612
| | - Deena Bader
- James A. Haley Veterans Hospital, Tampa, Florida 33612
| | - Rekha Patel
- Department of Chemistry, University of South Florida, Tampa, Florida 33612
| | - Michel Murr
- Surgery Department, University of Central Florida, Orlando, Florida 32816.,Bariatric and Metabolic Institute, AdventHealth, Tampa, Florida 33612
| | - Wayne Guida
- Department of Chemistry, University of South Florida, Tampa, Florida 33612
| | - Rutilio Fratti
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Illinois 61801
| | - Niketa A Patel
- Department of Molecular Medicine, University of South Florida, Tampa, Florida 33612 .,James A. Haley Veterans Hospital, Tampa, Florida 33612
| |
Collapse
|
15
|
Shi Y, Parag S, Patel R, Lui A, Murr M, Cai J, Patel NA. Stabilization of lncRNA GAS5 by a Small Molecule and Its Implications in Diabetic Adipocytes. Cell Chem Biol 2019; 26:319-330.e6. [PMID: 30661991 PMCID: PMC10498384 DOI: 10.1016/j.chembiol.2018.11.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/08/2018] [Accepted: 11/16/2018] [Indexed: 12/22/2022]
Abstract
Long noncoding RNA (lncRNA) are regulatory RNAs >200 nt. We previously showed that lncRNA GAS5 decreases significantly in serum of type 2 diabetes mellitus (T2DM) patients. Hence, we sought to decipher the molecular mechanisms underlying the role of GAS5 in T2DM in adipose tissue. Using CHIP-RIP, we demonstrate that GAS5 binds to promoter of insulin receptor to regulate its expression, and its depletion inhibits glucose uptake and insulin signaling. Toward stabilizing GAS5 levels in T2DM, we incorporated a strategy to limit the degradation of GAS5 by blocking the interaction of GAS5 and UPF1 with a small molecule identified using OBTC screening strategy. NP-C86 binds to GAS5 with high affinity, and increases GAS5 levels and glucose uptake in diabetic patient adipocytes. As a broader impact, NP-C86 may be used as a molecular probe to investigate the intricacies of GAS5 in relevant biological systems as it offers specificity, efficient cellular uptake and is non-cytotoxic.
Collapse
Affiliation(s)
- Yan Shi
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL 33620, USA
| | - Sajan Parag
- Department of Molecular Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | - Rekha Patel
- Department of Molecular Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | - Ashley Lui
- Department of Molecular Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | - Michel Murr
- Department of Surgery, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL 33620, USA.
| | - Niketa A Patel
- James A. Haley Veterans Hospital, 13000 Bruce B. Downs Boulevard, Tampa, FL 33612, USA; Department of Molecular Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA.
| |
Collapse
|
16
|
Stojanović S, Najman S, Korać A. Stem Cells Derived from Lipoma and Adipose Tissue-Similar Mesenchymal Phenotype but Different Differentiation Capacity Governed by Distinct Molecular Signature. Cells 2018; 7:E260. [PMID: 30544806 PMCID: PMC6316974 DOI: 10.3390/cells7120260] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
Lipomas are benign adipose tissue tumors of unknown etiology, which can vary in size, number, body localization and cell populations within the tissue. Lipoma-derived stem cells (LDSCs) are proposed as a potential tool in regenerative medicine and tissue engineering due to their similar characteristics with adipose-derived stem cells (ADSCs) reported so far. Our study is among the first giving detailed insights into the molecular signature and differences in the differentiation capacity of LDSCs in vitro compared to ADSCs. Mesenchymal stem cell phenotype was analyzed by gene expression and flow cytometric analysis of stem cell markers. Adipogenesis and osteogenesis were analyzed by microscopic analysis, cytochemical and immunocytochemical staining, gene and protein expression analyses. We showed that both LDSCs and ADSCs were mesenchymal stem cells with similar phenotype and stemness state but different molecular basis for potential differentiation. Adipogenesis-related genes expression pattern and presence of more mature adipocytes in ADSCs than in LDSCs after 21 days of adipogenic differentiation, indicated that differentiation capacity of LDSCs was significantly lower compared to ADSCs. Analysis of osteogenesis-related markers after 16 days of osteogenic differentiation revealed that both types of cells had characteristic osteoblast-like phenotype, but were at different stages of osteogenesis. Differences observed between LDSCs and ADSCs are probably due to the distinct molecular signature and their commitment in the tissue that governs their different capacity and fate during adipogenic and osteogenic induction in vitro despite their similar mesenchymal phenotype.
Collapse
Affiliation(s)
- Sanja Stojanović
- Department of Biology and Human Genetics and Department for Cell and Tissue Engineering, Faculty of Medicine, University of Niš, 18000 Niš, Serbia.
| | - Stevo Najman
- Department of Biology and Human Genetics and Department for Cell and Tissue Engineering, Faculty of Medicine, University of Niš, 18000 Niš, Serbia.
| | - Aleksandra Korać
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia.
| |
Collapse
|
17
|
Haney NM, Gabrielson A, Kohn TP, Hellstrom WJG. The Use of Stromal Vascular Fraction in the Treatment of Male Sexual Dysfunction: A Review of Preclinical and Clinical Studies. Sex Med Rev 2018; 7:313-320. [PMID: 29960873 DOI: 10.1016/j.sxmr.2018.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Stem cell therapy using stromal vascular fraction (SVF) is a promising treatment modality. SVF is comprised of a mixture of adipose-derived stem cells, endothelial precursor cells, and immune modulatory cells that act synergistically to facilitate angiogenesis and epithelial cell differentiation. This makes SVF an attractive option for men's sexual disorders that require reconstitution of vasculature and endothelial lining, namely erectile dysfunction (ED) and Peyronie's disease (PD). AIM The objective of this study was to compare and contrast the available literature regarding the use of SVF in the treatment of male sexual dysfunction. METHODS A literature review was performed in PubMed with the keywords "stromal vascular fraction" and/or "erectile dysfunction" and/or "Peyronie's disease" and/or "sexual dysfunction." MAIN OUTCOME MEASURES The main outcome measure for preclinical studies was erectile function, as measured by changes in intracavernous pressures, and results of histopathologic analysis of corporal tissue. Clinical endpoint analysis in humans included various patient questionnaires. RESULTS For ED, there were 5 preclinical studies included in the analysis, with 1 Phase 1 clinical trial in humans. Major limitations of both the preclinical and clinical studies included the absence of SVF component analysis, and short duration of follow-up. Despite a paucity of preclinical studies, there was a single clinical study assessing the efficacy of combination SVF and shock wave therapy in the treatment of PD. Limitations of this study included an absence of a control group and the use of subjective data. CONCLUSION Preclinical and clinical data in the use of SVF for the treatment of male sexual dysfunction is deficient. Even though multiple medicinal disciplines are studying the use of SVF on a myriad of pathologies, further investigative work elucidating the mechanism and potential adverse effects of SVF need to be performed before clinical trials are undertaken. Haney NM, Gabrielson A, Kohn TP, Hellstrom WJG. The Use of Stromal Vascular Fraction in the Treatment of Male Sexual Dysfunction: A Review of Preclinical and Clinical Studies. Sex Med Rev 2019;7:313-320.
Collapse
Affiliation(s)
- Nora M Haney
- Tulane University School of Medicine, Department of Urology, New Orleans, LA, USA
| | - Andrew Gabrielson
- Tulane University School of Medicine, Department of Urology, New Orleans, LA, USA
| | - Taylor P Kohn
- Baylor College of Medicine, Department of Urology, Houston, TX, USA
| | - Wayne J G Hellstrom
- Tulane University School of Medicine, Department of Urology, New Orleans, LA, USA.
| |
Collapse
|
18
|
Kishimoto N, Honda Y, Momota Y, Tran SD. Dedifferentiated Fat (DFAT) cells: A cell source for oral and maxillofacial tissue engineering. Oral Dis 2018; 24:1161-1167. [PMID: 29356251 DOI: 10.1111/odi.12832] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 12/26/2022]
Abstract
Tissue engineering is a promising method for the regeneration of oral and maxillofacial tissues. Proper selection of a cell source is important for the desired application. This review describes the discovery and usefulness of dedifferentiated fat (DFAT) cells as a cell source for tissue engineering. Dedifferentiated Fat cells are a highly homogeneous cell population (high purity), highly proliferative, and possess a multilineage potential for differentiation into various cell types under proper in vitro inducing conditions and in vivo. Moreover, DFAT cells have a higher differentiation capability of becoming osteoblasts, chondrocytes, and adipocytes than do bone marrow-derived mesenchymal stem cells and/or adipose tissue-derived stem cells. The usefulness of DFAT cells in vivo for periodontal tissue, bone, peripheral nerve, muscle, cartilage, and fat tissue regeneration was reported. Dedifferentiated Fat cells obtained from the human buccal fat pad (BFP) are a minimally invasive procedure with limited esthetic complications for patients. The BFP is a convenient and accessible anatomical site to harvest DFAT cells for dentists and oral surgeons, and thus is a promising cell source for oral and maxillofacial tissue engineering.
Collapse
Affiliation(s)
- N Kishimoto
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Y Honda
- Institute of Dental Research, Osaka Dental University, Osaka, Japan
| | - Y Momota
- Department of Anesthesiology, Osaka Dental University, Osaka, Japan
| | - S D Tran
- Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
19
|
Tsurumachi N, Akita D, Kano K, Matsumoto T, Toriumi T, Kazama T, Oki Y, Saito-Tamura Y, Tonogi M, Shimizu N, Honda M. Effect of collagenase concentration on the isolation of small adipocytes from human buccal fat pad. J Oral Sci 2018; 60:14-23. [PMID: 29479028 DOI: 10.2334/josnusd.16-0786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Dedifferentiated fat (DFAT) cells were isolated from mature adipocytes using the ceiling culture method. Recently, we successfully isolated DFAT cells from adipocytes with a relatively small size (<40 μm). DFAT cells have a higher osteogenic potential than that of medium adipocytes. Therefore, the objective of this study was to determine the optimal concentration of collagenase solution for isolating small adipocytes from human buccal fat pads (BFPs). Four concentrations of collagenase solution (0.01%, 0.02%, 0.1%, and 0.5%) were used, and their effectiveness was assessed by the number of small adipocytes and DFAT cells isolated. The total number of floating adipocytes that dissociated with 0.02% collagenase was 2.5 times of that dissociated with 0.1% collagenase. The number of floating adipocytes with a diameter of ≤29 μm that dissociated with 0.02% collagenase was thrice of those dissociated with 0.1% and 0.5% collagenase. The number of DFAT cells that dissociated with 0.02% collagenase was 1.5 times of that dissociated with 0.1% collagenase. In addition, DFAT cells that dissociated with 0.02% collagenase had a higher osteogenic differentiation potential than those that dissociated with 0.1% collagenase. These results suggest that 0.02% is the optimal collagenase concentration for isolating small adipocytes from BFPs.
Collapse
Affiliation(s)
- Niina Tsurumachi
- Department of Orthodontics, Nihon University School of Dentistry
| | - Daisuke Akita
- Department of Partial Denture Prosthodontics, Nihon University School of Dentistry
| | - Koichiro Kano
- Laboratory of Cell and Tissue Biology, College of Bioresource Sciences, Nihon University
| | - Taro Matsumoto
- Department of Functional Morphology Division of Cell Regeneration and Transplantation, Nihon University School of Medicine
| | - Taku Toriumi
- Department of Anatomy, Nihon University School of Dentistry
| | - Tomohiko Kazama
- Department of Functional Morphology Division of Cell Regeneration and Transplantation, Nihon University School of Medicine
| | - Yoshinao Oki
- Laboratory of Cell and Tissue Biology, College of Bioresource Sciences, Nihon University
| | | | - Morio Tonogi
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry
| | | | - Masaki Honda
- Department of Oral Anatomy, Aichi Gakuin University School of Dentistry
| |
Collapse
|
20
|
Impact of Antibiotics on the Proliferation and Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2017; 18:ijms18122522. [PMID: 29186789 PMCID: PMC5751125 DOI: 10.3390/ijms18122522] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a promising source of mesenchymal stem cells. Their potential to differentiate and regenerate other types of tissues may be affected by several factors. This may be due to in vitro cell-culture conditions, especially the supplementation with antibiotics. The aim of our study was to evaluate the effects of a penicillin-streptomycin mixture (PS), amphotericin B (AmB), a complex of AmB with copper (II) ions (AmB-Cu2+) and various combinations of these antibiotics on the proliferation and differentiation of adipose-derived stem cells in vitro. Normal human adipose-derived stem cells (ADSC, Lonza) were routinely maintained in a Dulbecco’s Modified Eagle Medium (DMEM) that was either supplemented with selected antibiotics or without antibiotics. The ADSC that were used for the experiment were at the second passage. The effect of antibiotics on proliferation was analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and sulforhodamine-B (SRB) tests. Differentiation was evaluated based on Alizarin Red staining, Oil Red O staining and determination of the expression of ADSC, osteoblast and adipocyte markers by real-time RT-qPCR. The obtained results indicate that the influence of antibiotics on adipose-derived stem cells depends on the duration of exposure and on the combination of applied compounds. We show that antibiotics alter the proliferation of cells and also promote natural osteogenesis, and adipogenesis, and that this effect is also noticeable in stimulated osteogenesis.
Collapse
|
21
|
El Bassit G, Patel RS, Carter G, Shibu V, Patel AA, Song S, Murr M, Cooper DR, Bickford PC, Patel NA. MALAT1 in Human Adipose Stem Cells Modulates Survival and Alternative Splicing of PKCδII in HT22 Cells. Endocrinology 2017; 158:183-195. [PMID: 27841943 PMCID: PMC5412980 DOI: 10.1210/en.2016-1819] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/04/2016] [Indexed: 01/15/2023]
Abstract
Brain injury may be caused by trauma or may occur in stroke and neurodegenerative diseases. Because the central nervous system is unable to regenerate efficiently, there is utmost interest in the use of stem cells to promote neuronal survival. Of interest here are human adipose-derived stem cells (hASCs), which secrete factors that enhance regeneration and survival of neurons in sites of injury. We evaluated the effect of hASC secretome on immortalized mouse hippocampal cell line (HT22) after injury. Protein kinase C δ (PKCδ) activates survival and proliferation in neurons and is implicated in memory. We previously showed that alternatively spliced PKCδII enhances neuronal survival via B-cell lymphoma 2 Bcl2 in HT22 neuronal cells. Our results demonstrate that following injury, treatment with exosomes from the hASC secretome increases expression of PKCδII in HT22 cells and increases neuronal survival and proliferation. Specifically, we demonstrate that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long noncoding RNA contained in the hASC exosomes mediates PKCδII splicing, thereby increasing neuronal survival. Using antisense oligonucleotides for MALAT1 and RNA immunoprecipitation assays, we demonstrate that MALAT1 recruits splice factor serine-arginine-rich splice factor 2 (SRSF2) to promote alternative splicing of PKCδII. Finally, we evaluated the role of insulin in enhancing hASC-mediated neuronal survival and demonstrated that insulin treatment dramatically increases the association of MALAT1 and SRSF2 and substantially increases survival and proliferation after injury in HT22 cells. In conclusion, we demonstrate the mechanism of action of hASC exosomes in increasing neuronal survival. This effect of hASC exosomes to promote wound healing can be further enhanced by insulin treatment in HT22 cells.
Collapse
Affiliation(s)
| | | | - Gay Carter
- James A. Haley Veterans Hospital, Tampa, Florida 33612; and
| | | | | | - Shijie Song
- James A. Haley Veterans Hospital, Tampa, Florida 33612; and
| | | | - Denise R. Cooper
- James A. Haley Veterans Hospital, Tampa, Florida 33612; and
- Molecular Medicine,
| | - Paula C. Bickford
- James A. Haley Veterans Hospital, Tampa, Florida 33612; and
- Neurosurgery and Brain Survival, University of South Florida, Tampa, Florida 33612
| | - Niketa A. Patel
- James A. Haley Veterans Hospital, Tampa, Florida 33612; and
- Molecular Medicine,
| |
Collapse
|
22
|
Mesenchymal Stem Cells as a Prospective Therapy for the Diabetic Foot. Stem Cells Int 2016; 2016:4612167. [PMID: 27867398 PMCID: PMC5102750 DOI: 10.1155/2016/4612167] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 12/14/2022] Open
Abstract
The diabetic foot is a serious complication of diabetes. Mesenchymal stem cells are an abundant source of stem cells which occupy a special position in cell therapies, and recent studies have suggested that mesenchymal stem cells can play essential roles in treatments for the diabetic foot. Here, we discuss the advances that have been made in mesenchymal stem cell treatments for this condition. The roles and functional mechanisms of mesenchymal stem cells in the diabetic foot are also summarized, and insights into current and future studies are presented.
Collapse
|
23
|
SOX2, OCT3/4 and NANOG expression and cellular plasticity in rare human somatic cells requires CD73. Cell Signal 2016; 28:1923-1932. [PMID: 27705752 DOI: 10.1016/j.cellsig.2016.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/24/2016] [Indexed: 02/06/2023]
Abstract
Endogenous Plastic Somatic (ePS) cells isolated from adult human tissues exhibit extensive lineage plasticity in vitro and in vivo. Here we visualize these rare ePS cells in a latent state, i.e. lacking SOX2, OCT3/4 and NANOG (SON) expression, in non-diseased breast specimens through immunohistochemical analysis of previously identified ePS-specific biomarkers (CD73+, EpCAM+ and CD90-). We also report a novel mechanism by which these latent ePS cells acquire SON expression and plasticity in vitro. Four extracellular factors are necessary for the acquisition of SON expression and lineage plasticity in ePS cells: adenosine (which is produced by the 5' ecto-nucleotidase CD73 and activates in turn the PKA-dependent IL6/STAT3 pathway through the adenosine receptor ADORA2b), IL6, FGF2 and ACTIVIN A. Blocking any pathway component renders ePS cells incapable of SON expression and lineage plasticity. Notably, hESCs do not use adenosine or IL6 nor they express CD73 or ADORA2b and inhibition of adenosine signaling does not ablate their plasticity. Therefore, the data presented here delineate novel circuitry and physiological signals for accessing SON expression in rare, undifferentiated human cells.
Collapse
|
24
|
Patel RS, Carter G, El Bassit G, Patel AA, Cooper DR, Murr M, Patel NA. Adipose-derived stem cells from lean and obese humans show depot specific differences in their stem cell markers, exosome contents and senescence: role of protein kinase C delta (PKCδ) in adipose stem cell niche. Stem Cell Investig 2016; 3:2. [PMID: 27358894 DOI: 10.3978/j.issn.2306-9759.2016.01.02] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 01/21/2016] [Indexed: 01/23/2023]
Abstract
BACKGROUND Adipose-derived stem cells (ASC) and its exosomes are gaining utmost importance in the field of regenerative medicine. The ASCs tested for their potential in wound healing are predominantly derived from the subcutaneous depot of lean donors. However, it is important to characterize the ASC derived from different adipose depots as these depots have clinically distinct roles. METHODS We characterized the ASC derived from subcutaneous and omental depots from a lean donor (sc-ASCn and om-ASCn) and compared it to the ASC derived from an obese donor (sc-ASCo and om-ASCo) using flow cytometry and real time qPCR. RESULTS We show that stem cell markers Oct4, Sal4, Sox15, KLF4 and BMI1 have distinct expression patterns in each ASC. We evaluated the secretome of the ASC and characterized their secreted exosomes. We show long noncoding RNAs (lncRNAs) are secreted by ASC and their expression varied between the ASC's derived from different depots. Protein kinase C delta (PKCδ) regulates the mitogenic signals in stem cells. We evaluated the effect of silencing PKCδ in sc-ASCn, om-ASCn, sc-ASCo and om-ASCo. Using β-galactosidase staining, we evaluated the percentage of senescent cells in sc-ASCn, om-ASCn, sc-ASCo and om-ASCo. Our results also indicated that silencing PKCδ increases the percentage of senescent cells. CONCLUSIONS Our case-specific study demonstrates a role of PKCδ in maintaining the adipose stem cell niche and importantly demonstrates depot-specific differences in adipose stem cells and their exosome content.
Collapse
Affiliation(s)
- Rekha S Patel
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Gay Carter
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Ghattas El Bassit
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Achintya A Patel
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Denise R Cooper
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Michel Murr
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Niketa A Patel
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| |
Collapse
|
25
|
Huber B, Kluger PJ. Decelerating Mature Adipocyte Dedifferentiation by Media Composition. Tissue Eng Part C Methods 2015; 21:1237-45. [DOI: 10.1089/ten.tec.2015.0166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Birgit Huber
- Institute of Interfacial Process Engineering and Plasma Technology, University of Stuttgart, Stuttgart, Germany
| | - Petra J. Kluger
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Process Analysis & Technology (PA&T), Reutlingen University, Reutlingen, Germany
| |
Collapse
|
26
|
Jumabay M, Boström KI. Dedifferentiated fat cells: A cell source for regenerative medicine. World J Stem Cells 2015; 7:1202-1214. [PMID: 26640620 PMCID: PMC4663373 DOI: 10.4252/wjsc.v7.i10.1202] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/02/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
The identification of an ideal cell source for tissue regeneration remains a challenge in the stem cell field. The ability of progeny cells to differentiate into other cell types is important for the processes of tissue reconstruction and tissue engineering and has clinical, biochemical or molecular implications. The adaptation of stem cells from adipose tissue for use in regenerative medicine has created a new role for adipocytes. Mature adipocytes can easily be isolated from adipose cell suspensions and allowed to dedifferentiate into lipid-free multipotent cells, referred to as dedifferentiated fat (DFAT) cells. Compared to other adult stem cells, the DFAT cells have unique advantages in their abundance, ease of isolation and homogeneity. Under proper condition in vitro and in vivo, the DFAT cells have exhibited adipogenic, osteogenic, chondrogenic, cardiomyogenc, angiogenic, myogenic, and neurogenic potentials. In this review, we first discuss the phenomena of dedifferentiation and transdifferentiation of cells, and then dedifferentiation of adipocytes in particular. Understanding the dedifferentiation process itself may contribute to our knowledge of normal growth processes, as well as mechanisms of disease. Second, we highlight new developments in DFAT cell culture and summarize the current understanding of DFAT cell properties. The unique features of DFAT cells are promising for clinical applications such as tissue regeneration.
Collapse
|
27
|
Application of Green Tea Catechin for Inducing the Osteogenic Differentiation of Human Dedifferentiated Fat Cells in Vitro. Int J Mol Sci 2015; 16:27988-8000. [PMID: 26602917 PMCID: PMC4691028 DOI: 10.3390/ijms161226081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022] Open
Abstract
Despite advances in stem cell biology, there are few effective techniques to promote the osteogenic differentiation of human primary dedifferentiated fat (DFAT) cells. We attempted to investigate whether epigallocatechin-3-gallate (EGCG), the main component of green tea catechin, facilitates early osteogenic differentiation and mineralization on DFAT cells in vitro. DFAT cells were treated with EGCG (1.25-10 μM) in osteogenic medium (OM) with or without 100 nM dexamethasone (Dex) for 12 days (hereafter two osteogenic media were designated as OM(Dex) and OM). Supplementation of 1.25 μM EGCG to both the media effectively increased the mRNA expression of collagen 1 (COL1A1) and runt-related transcription factor 2 (RUNX2) and also increased proliferation and mineralization. Compared to OM(Dex) with EGCG, OM with EGCG induced earlier expression for COL1A1 and RUNX2 at day 1 and higher mineralization level at day 12. OM(Dex) with 10 μM EGCG remarkably hampered the proliferation of the DFAT cells. These results suggest that OM(without Dex) with EGCG might be a preferable medium to promote proliferation and to induce osteoblast differentiation of DFAT cells. Our findings provide an insight for the combinatory use of EGCG and DFAT cells for bone regeneration and stem cell-based therapy.
Collapse
|
28
|
Stromal vascular fraction: A regenerative reality? Part 2: Mechanisms of regenerative action. J Plast Reconstr Aesthet Surg 2015; 69:180-8. [PMID: 26546112 DOI: 10.1016/j.bjps.2015.10.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/26/2015] [Accepted: 10/13/2015] [Indexed: 12/22/2022]
Abstract
Adipose tissue is a rich source of cells with emerging promise for tissue engineering and regenerative medicine. The stromal vascular fraction (SVF), in particular, is an eclectic composite of cells with progenitor activity that includes preadipocytes, mesenchymal stem cells, pericytes, endothelial cells, and macrophages. SVF has enormous potential for therapeutic application and is being investigated for multiple clinical indications including lipotransfer, diabetes-related complications, nerve regeneration, burn wounds and numerous others. In Part 2 of our review, we explore the basic science behind the regenerative success of the SVF and discuss significant mechanisms that are at play. The existing literature suggests that angiogenesis, immunomodulation, differentiation, and extracellular matrix secretion are the main avenues through which regeneration and healing is achieved by the stromal vascular fraction.
Collapse
|
29
|
Wollina U. Midfacial rejuvenation by hyaluronic acid fillers and subcutaneous adipose tissue – A new concept. Med Hypotheses 2015; 84:327-30. [DOI: 10.1016/j.mehy.2015.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 11/27/2022]
|
30
|
Lessard J, Côté JA, Lapointe M, Pelletier M, Nadeau M, Marceau S, Biertho L, Tchernof A. Generation of human adipose stem cells through dedifferentiation of mature adipocytes in ceiling cultures. J Vis Exp 2015:52485. [PMID: 25867041 PMCID: PMC4401230 DOI: 10.3791/52485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mature adipocytes have been shown to reverse their phenotype into fibroblast-like cells in vitro through a technique called ceiling culture. Mature adipocytes can also be isolated from fresh adipose tissue for depot-specific characterization of their function and metabolic properties. Here, we describe a well-established protocol to isolate mature adipocytes from adipose tissues using collagenase digestion, and subsequent steps to perform ceiling cultures. Briefly, adipose tissues are incubated in a Krebs-Ringer-Henseleit buffer containing collagenase to disrupt tissue matrix. Floating mature adipocytes are collected on the top surface of the buffer. Mature cells are plated in a T25-flask completely filled with media and incubated upside down for a week. An alternative 6-well plate culture approach allows the characterization of adipocytes undergoing dedifferentiation. Adipocyte morphology drastically changes over time of culture. Immunofluorescence can be easily performed on slides cultivated in 6-well plates as demonstrated by FABP4 immunofluorescence staining. FABP4 protein is present in mature adipocytes but down-regulated through dedifferentiation of fat cells. Mature adipocyte dedifferentiation may represent a new avenue for cell therapy and tissue engineering.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - André Tchernof
- IUCPQ Research Center; CHU de Québec Research Center; Laval University;
| |
Collapse
|
31
|
Kaku M, Akiba Y, Akiyama K, Akita D, Nishimura M. Cell-based bone regeneration for alveolar ridge augmentation--cell source, endogenous cell recruitment and immunomodulatory function. J Prosthodont Res 2015; 59:96-112. [PMID: 25749435 DOI: 10.1016/j.jpor.2015.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/05/2015] [Indexed: 11/30/2022]
Abstract
Alveolar ridge plays a pivotal role in supporting dental prosthesis particularly in edentulous and semi-dentulous patients. However the alveolar ridge undergoes atrophic change after tooth loss. The vertical and horizontal volume of the alveolar ridge restricts the design of dental prosthesis; thus, maintaining sufficient alveolar ridge volume is vital for successful oral rehabilitation. Recent progress in regenerative approaches has conferred marked benefits in prosthetic dentistry, enabling regeneration of the atrophic alveolar ridge. In order to achieve successful alveolar ridge augmentation, sufficient numbers of osteogenic cells are necessary; therefore, autologous osteoprogenitor cells are isolated, expanded in vitro, and transplanted to the specific anatomical site where the bone is required. Recent studies have gradually elucidated that transplanted osteoprogenitor cells are not only a source of bone forming osteoblasts, they appear to play multiple roles, such as recruitment of endogenous osteoprogenitor cells and immunomodulatory function, at the forefront of bone regeneration. This review focuses on the current consensus of cell-based bone augmentation therapies with emphasis on cell sources, transplanted cell survival, endogenous stem cell recruitment and immunomodulatory function of transplanted osteoprogenitor cells. Furthermore, if we were able to control the mobilization of endogenous osteoprogenitor cells, large-scale surgery may no longer be necessary. Such treatment strategy may open a new era of safer and more effective alveolar ridge augmentation treatment options.
Collapse
Affiliation(s)
- Masaru Kaku
- Division of Bioprosthodontics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Yosuke Akiba
- Division of Bioprosthodontics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kentaro Akiyama
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Daisuke Akita
- Department of Partial Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Masahiro Nishimura
- Department of Oral Maxillofacial Prosthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
32
|
Limbourg A, Schnabel S, Lozanovski VJ, Napp LC, Ha TC, Maetzig T, Bauersachs J, Naim HY, Schambach A, Limbourg FP. Genetic reporter analysis reveals an expandable reservoir of OCT4+ cells in adult skin. ACTA ACUST UNITED AC 2014; 3:9. [PMID: 25408888 PMCID: PMC4230759 DOI: 10.1186/2045-9769-3-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 06/03/2014] [Indexed: 12/31/2022]
Abstract
The transcription factor Oct4 (Pou5f1) is a critical regulator of pluripotency in embryonic and induced pluripotent stem cells. Therefore, Oct4 expression might identify somatic stem cell populations with inherent multipotent potential or a propensity for facilitated reprogramming. However, analysis of Oct4 expression is confounded by Oct4 pseudogenes or non-pluripotency-related isoforms. Systematic analysis of a transgenic Oct4-EGFP reporter mouse identified testis and skin as two principle sources of Oct4+ cells in postnatal mice. While the prevalence of GFP+ cells in testis rapidly declined with age, the skin-resident GFP+ population expanded in a cyclical fashion. These cells were identified as epidermal stem cells dwelling in the stem cell niche of the hair follicle, which endogenously expressed all principle reprogramming factors at low levels. Interestingly, skin wounding or non-traumatic hair removal robustly expanded the GFP+ epidermal cell pool not only locally, but also in uninjured skin areas, demonstrating the existence of a systemic response. Thus, the epithelial stem cell niche of the hair follicle harbors an expandable pool of Oct4+ stem cells, which might be useful for therapeutic cell transfer or facilitated reprogramming.
Collapse
Affiliation(s)
- Anne Limbourg
- Research Group Regenerative Agents, Hannover, Germany ; REBIRTH Cluster of Excellence, Hannover, Germany ; Integrated Research Center Transplantation (IFB-Tx), Hannover, Germany ; Department of Plastic, Hand and Reconstructive Surgery, Hannover, Germany
| | - Sabine Schnabel
- Research Group Regenerative Agents, Hannover, Germany ; REBIRTH Cluster of Excellence, Hannover, Germany
| | - Vladimir J Lozanovski
- Research Group Regenerative Agents, Hannover, Germany ; REBIRTH Cluster of Excellence, Hannover, Germany ; Department of General and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - L Christian Napp
- Research Group Regenerative Agents, Hannover, Germany ; Department of Cardiology and Angiology, Hannover, Germany
| | - Teng-Cheong Ha
- REBIRTH Cluster of Excellence, Hannover, Germany ; Institute of Experimental Hematology, OE6960 Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Tobias Maetzig
- REBIRTH Cluster of Excellence, Hannover, Germany ; Institute of Experimental Hematology, OE6960 Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Johann Bauersachs
- REBIRTH Cluster of Excellence, Hannover, Germany ; Department of Cardiology and Angiology, Hannover, Germany
| | - Hassan Y Naim
- Department of Physiological Chemistry, Hannover Veterinary School, Hannover, Germany
| | - Axel Schambach
- REBIRTH Cluster of Excellence, Hannover, Germany ; Integrated Research Center Transplantation (IFB-Tx), Hannover, Germany ; Institute of Experimental Hematology, OE6960 Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany ; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Harvard, USA
| | - Florian P Limbourg
- Research Group Regenerative Agents, Hannover, Germany ; REBIRTH Cluster of Excellence, Hannover, Germany ; Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany ; Vascular Medicine and Transplantation Research, Dept. of Nephrology and Hypertension, OE 6841, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
33
|
Carter G, Patel R, Apostolatos A, Murr M, Cooper DR, Patel NA. Protein kinase C delta (PKCδ) splice variant modulates senescence via hTERT in adipose-derived stem cells. Stem Cell Investig 2014; 1:3. [PMID: 27358850 DOI: 10.3978/j.issn.2306-9759.2014.01.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/17/2014] [Indexed: 11/14/2022]
Abstract
BACKGROUND Adipose-derived stem cells (ADSC) were isolated and characterized from lean and obese subjects. We previously reported that distinct differences were observed in differentiating lean and obese preadipocytes. Protein kinase C delta (PKCδ) is alternatively spliced and has important roles in apoptosis. PKCδI promotes apoptosis and PKCδVIII promotes survival. Our previous data indicated an increase in the survival kinase, PKCδVIII in ADSC derived from an obese donor. We also determined that obese adipocytes were resistant to apoptosis. Here, we determine the relationship between a survival kinase PKCδVIII and hTERT expression in adipose derived stem cells from a lean and obese subject. METHODS We evaluated the telomerase activity and human telomerase reverse transcriptase (hTERT) expression in lean and obese ADSC. The lean and obese ADSC were purchased as cryopreserved cells from ZenBio™ (Research Triangle Park, NC, USA). Analyses were performed using PRISM™ software and analyzed using two-tailed Student's t-test. RESULTS We observed an increase in telomerase in differentiating obese ADSC using western blot analysis. We determined the levels of hTERT splice variants. hTERT α+/β+ splice variant was increased after transfected of PKCδVIII. We next determined whether PKCδVIII over-expression affected the levels of telomerase. The results indicate an increase in telomerase with PKCδVIII over-expression. CONCLUSIONS Over-expression of PKCδVIII in lean ADSC substantially increased expression of hTERT and telomerase. The decreased senescence seen in obese ADSC may in part be attributed to PKCδVIII. Obese ADSC undergo lower senescence and may have increased growth potential. These results propose a larger epigenetic modification in obese ADSC compared to lean ADSC.
Collapse
Affiliation(s)
- Gay Carter
- 1 James A Haley Veterans Hospital, Tampa, FL, USA ; 2 Department of Molecular Medicine and 3 Surgery, University of South Florida, Tampa, FL, USA
| | - Rekha Patel
- 1 James A Haley Veterans Hospital, Tampa, FL, USA ; 2 Department of Molecular Medicine and 3 Surgery, University of South Florida, Tampa, FL, USA
| | - André Apostolatos
- 1 James A Haley Veterans Hospital, Tampa, FL, USA ; 2 Department of Molecular Medicine and 3 Surgery, University of South Florida, Tampa, FL, USA
| | - Michel Murr
- 1 James A Haley Veterans Hospital, Tampa, FL, USA ; 2 Department of Molecular Medicine and 3 Surgery, University of South Florida, Tampa, FL, USA
| | - Denise R Cooper
- 1 James A Haley Veterans Hospital, Tampa, FL, USA ; 2 Department of Molecular Medicine and 3 Surgery, University of South Florida, Tampa, FL, USA
| | - Niketa A Patel
- 1 James A Haley Veterans Hospital, Tampa, FL, USA ; 2 Department of Molecular Medicine and 3 Surgery, University of South Florida, Tampa, FL, USA
| |
Collapse
|