1
|
Sun B, Meng XH, Li YM, Lin H, Xiao ZD. MicroRNA-18a prevents senescence of mesenchymal stem cells by targeting CTDSPL. Aging (Albany NY) 2024; 16:4904-4919. [PMID: 38460957 PMCID: PMC10968691 DOI: 10.18632/aging.205642] [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] [Received: 08/29/2023] [Accepted: 12/26/2023] [Indexed: 03/11/2024]
Abstract
Stem cell therapy requires massive-scale homogeneous stem cells under strict qualification control. However, Prolonged ex vivo expansion impairs the biological functions and results in senescence of mesenchymal stem cells (MSCs). We investigated the function of CTDSPL in the premature senescence process of MSCs and clarified that miR-18a-5p played a prominent role in preventing senescence of long-term cultured MSCs and promoting the self-renewal ability of MSCs. Over-expression of CTDSPL resulted in an enlarged morphology, up-regulation of p16 and accumulation of SA-β-gal of MSCs. The reduced phosphorylated RB suggested cell cycle arrest of MSCs. All these results implied that CTDSPL induced premature senescence of MSCs. We further demonstrated that miR-18a-5p was a putative regulator of CTDSPL by luciferase reporter assay. Inhibition of miR-18a-5p promoted the expression of CTDSPL and induced premature senescence of MSCs. Continuous overexpression of miR-18a-5p improved self-renewal of MSCs by reducing ROS level, increased expression of Oct4 and Nanog, and promoted growth rate and differentiation capability. We reported for the first time that the dynamic interaction of miR-18a-5p and CTDSPL is crucial for stem cell senescence.
Collapse
Affiliation(s)
- Bo Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xian-Hui Meng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yu-Min Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Hao Lin
- Department of Clinical Science and Research, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhong-Dang Xiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
2
|
Alessio N, Aprile D, Peluso G, Mazzone V, Patrone D, Di Bernardo G, Galderisi U. IGFBP5 is released by senescent cells and is internalized by healthy cells, promoting their senescence through interaction with retinoic receptors. Cell Commun Signal 2024; 22:122. [PMID: 38351010 PMCID: PMC10863175 DOI: 10.1186/s12964-024-01469-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
Cells that are exposed to harmful genetic damage, either from internal or external sources, may undergo senescence if they are unable to repair their DNA. Senescence, characterized by a state of irreversible growth arrest, can spread to neighboring cells through a process known as the senescence-associated secretory phenotype (SASP). This phenomenon contributes to both aging and the development of cancer. The SASP comprises a variety of factors that regulate numerous functions, including the induction of secondary senescence, modulation of immune system activity, remodeling of the extracellular matrix, alteration of tissue structure, and promotion of cancer progression. Identifying key factors within the SASP is crucial for understanding the underlying mechanisms of senescence and developing effective strategies to counteract cellular senescence. Our research has specifically focused on investigating the role of IGFBP5, a component of the SASP observed in various experimental models and conditions.Through our studies, we have demonstrated that IGFBP5 actively contributes to promoting senescence and can induce senescence in neighboring cells. We have gained valuable insights into the mechanisms through which IGFBP5 exerts its pro-senescence effects. These mechanisms include its release following genotoxic stress, involvement in signaling pathways mediated by reactive oxygen species and prostaglandins, internalization via specialized structures called caveolae, and interaction with a specific protein known as RARα. By uncovering these mechanisms, we have advanced our understanding of the intricate role of IGFBP5 in the senescence process. The significance of IGFBP5 as a pro-aging factor stems from an in vivo study we conducted on patients undergoing Computer Tomography analysis. In these patients, we observed an elevation in circulating IGFBP5 levels in response to radiation-induced organismal stress.Globally, our findings highlight the potential of IGFBP5 as a promising therapeutic target for age-related diseases and cancer.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, via Luigi De Crecchio 7, Naples, 80138, Italy
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, via Luigi De Crecchio 7, Naples, 80138, Italy
| | | | - Valeria Mazzone
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, via Luigi De Crecchio 7, Naples, 80138, Italy
| | - Deanira Patrone
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, via Luigi De Crecchio 7, Naples, 80138, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, via Luigi De Crecchio 7, Naples, 80138, Italy.
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, via Luigi De Crecchio 7, Naples, 80138, Italy.
- Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey.
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine Temple University, PA, Philadelphia, USA.
| |
Collapse
|
3
|
Jarrold BB, Tan CYR, Ho CY, Soon AL, Lam TT, Yang X, Nguyen C, Guo W, Chew YC, DeAngelis YM, Costello L, De Los Santos Gomez P, Przyborski S, Bellanger S, Dreesen O, Kimball AB, Oblong JE. Early onset of senescence and imbalanced epidermal homeostasis across the decades in photoexposed human skin: Fingerprints of inflammaging. Exp Dermatol 2022; 31:1748-1760. [DOI: 10.1111/exd.14654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/11/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - Chin Yee Ho
- A*STAR Skin Research Labs Singapore City Singapore
| | - Ai Ling Soon
- A*STAR Skin Research Labs Singapore City Singapore
| | - TuKiet T. Lam
- Keck MS & Proteomics Resource Yale School of Medicine New Haven Connecticut USA
| | | | | | - Wei Guo
- Zymo Research Corporation Irvine California USA
| | | | | | | | | | | | | | | | - Alexa B. Kimball
- Beth Israel Deaconess Medical Center and Harvard Medical School Boston Massachusetts USA
| | | |
Collapse
|
4
|
Feng Y, Luo J, Cheng J, Xu A, Qiu D, He S, Zheng D, Jia C, Zhang Q, Lin N. A Small-Molecule Cocktails-Based Strategy in Culture of Mesenchymal Stem Cells. Front Bioeng Biotechnol 2022; 10:819148. [PMID: 35360405 PMCID: PMC8963903 DOI: 10.3389/fbioe.2022.819148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/11/2022] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a variety of unique properties, such as stem cell multipotency and immune regulation, making them attractive for use in cell therapy. Before infusion therapy, MSCs are required to undergo tissue separation, purification, and expansion in vitro for a certain duration. During the process of in vitro expansion of MSCs, the influence of culture time and environment can lead to cell senescence, increased heterogeneity, and function attenuation, which limits their clinical applications. We used a cocktail of three small-molecule compounds, ACY (A-83–01, CHIR99021, and Y-27632), to increase the proliferation activity of MSCs in vitro and reduce cell senescence. ACY inhibited the increase in heterogeneity of MSCs and conserved their differentiation potential. Additionally, ACY maintained the phenotype of MSCs and upregulated the expression of immunomodulatory factors. These results suggest that ACY can effectively improve the quantity and quality of MSCs.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jintao Cheng
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Aimin Xu
- The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Dongbo Qiu
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Sixiao He
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dayong Zheng
- The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Changchang Jia
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| | - Qi Zhang
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| |
Collapse
|
5
|
Ueda S, Tominaga T, Ochi A, Sakurai A, Nishimura K, Shibata E, Wakino S, Tamaki M, Nagai K. TGF-β1 is involved in senescence-related pathways in glomerular endothelial cells via p16 translocation and p21 induction. Sci Rep 2021; 11:21643. [PMID: 34737348 PMCID: PMC8569175 DOI: 10.1038/s41598-021-01150-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
p16 inhibits cyclin-dependent kinases and regulates senescence-mediated arrest as well as p21. Nuclear p16 promotes G1 cell cycle arrest and cellular senescence. In various glomerular diseases, nuclear p16 expression is associated with disease progression. Therefore, the location of p16 is important. However, the mechanism of p16 trafficking between the nucleus and cytoplasm is yet to be fully investigated. TGF-β1, a major cytokine involved in the development of kidney diseases, can upregulate p21 expression. However, the relationship between TGF-β1 and p16 is poorly understood. Here, we report the role of podocyte TGF-β1 in regulating the p16 behavior in glomerular endothelial cells. We analyzed podocyte-specific TGF-β1 overexpression mice. Although p16 was found in the nuclei of glomerular endothelial cells and led to endothelial cellular senescence, the expression of p16 did not increase in glomeruli. In cultured endothelial cells, TGF-β1 induced nuclear translocation of p16 without increasing its expression. Among human glomerular diseases, p16 was detected in the nuclei of glomerular endothelial cells. In summary, we demonstrated the novel role of podocyte TGF-β1 in managing p16 behavior and cellular senescence in glomeruli, which has clinical relevance for the progression of human glomerular diseases.
Collapse
Affiliation(s)
- Sayo Ueda
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Arisa Ochi
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Akiko Sakurai
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Kenji Nishimura
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Eriko Shibata
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Shu Wakino
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Masanori Tamaki
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Kojiro Nagai
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan.
| |
Collapse
|
6
|
Che J, Xu C, Wu Y, Jia P, Han Q, Ma Y, Wang X, Du Y, Zheng Y. Early-senescent bone marrow mesenchymal stem cells promote C2C12 cell myogenic differentiation by preventing the nuclear translocation of FOXO3. Life Sci 2021; 277:119520. [PMID: 33887345 DOI: 10.1016/j.lfs.2021.119520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
AIMS Mouse bone marrow mesenchymal stem cells (BMSCs) are pluripotent cells with self-renewal and differentiation abilities. Since the effects of senescent BMSCs on C2C12 cells are not fully clear, the present study aimed to elucidate these effects. MAIN METHODS Senescence-associated β-galactosidase staining and western blotting were performed to confirm the senescence of BMSCs. Immunofluorescence and western blotting were used to assess myoblast differentiation in each group. The role of the AKT/P70 signaling pathway and forkhead box O3 (FOXO3) nuclear translocation was explored by western blotting. BMSC-derived exosomes were injected into the tibialis anterior of mice, and RT-qPCR was used to assess the role of exosomes in promoting muscle differentiation. KEY FINDINGS Conditioned medium (CM) from early-senescent BMSCs promoted myogenic differentiation in vitro, which was detected as enhanced expression of myosin heavy chain (MHC), myogenin (MYOG), and myogenic differentiation 1 (MyoD). The AKT signaling pathway was found to be regulated by CM, which inhibited FOXO3 nuclear translocation. RT-qPCR analysis results showed that MHC, MyoD, and MYOG mRNA expression increased in the tibialis anterior of mice after exosome injection. SIGNIFICANCE The present study demonstrated that early-senescent BMSCs accelerated C2C12 cell myogenic differentiation, and the transcription factor, FOXO3, was the target of senescent cells. Collectively, our results suggest that the AKT/P70 signaling pathway mediates the effect of BMSCs on neighboring cells.
Collapse
Affiliation(s)
- Ji Che
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China
| | - Cuidi Xu
- Department of Osteoporosis and Bone Disease, Huadong Hospital, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute, Shanghai, China
| | - Yuanyuan Wu
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China
| | - Peiyu Jia
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China
| | - Qi Han
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China
| | - Yantao Ma
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China
| | - Xiaolei Wang
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China
| | - Yijie Du
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China; Qingpu Traditional Chinese Medicine Hospital, Shanghai, China.
| | - Yongjun Zheng
- Department of Pain, Huadong Hospital, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Harada S, Mabuchi Y, Kohyama J, Shimojo D, Suzuki S, Kawamura Y, Araki D, Suyama T, Kajikawa M, Akazawa C, Okano H, Matsuzaki Y. FZD5 regulates cellular senescence in human mesenchymal stem/stromal cells. Stem Cells 2020; 39:318-330. [PMID: 33338299 PMCID: PMC7986096 DOI: 10.1002/stem.3317] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
Human mesenchymal stem/stromal cells (hMSCs) have garnered enormous interest as a potential resource for cell‐based therapies. However, the molecular mechanisms regulating senescence in hMSCs remain unclear. To elucidate these mechanisms, we performed gene expression profiling to compare clonal immature MSCs exhibiting multipotency with less potent MSCs. We found that the transcription factor Frizzled 5 (FZD5) is expressed specifically in immature hMSCs. The FZD5 cell surface antigen was also highly expressed in the primary MSC fraction (LNGFR+THY‐1+) and cultured MSCs. Treatment of cells with the FZD5 ligand WNT5A promoted their proliferation. Upon FZD5 knockdown, hMSCs exhibited markedly attenuated proliferation and differentiation ability. The observed increase in the levels of senescence markers suggested that FZD5 knockdown promotes cellular senescence by regulating the noncanonical Wnt pathway. Conversely, FZD5 overexpression delayed cell cycle arrest during the continued culture of hMSCs. These results indicated that the intrinsic activation of FZD5 plays an essential role in negatively regulating senescence in hMSCs and suggested that controlling FZD5 signaling offers the potential to regulate hMSC quality and improve the efficacy of cell‐replacement therapies using hMSCs.
Collapse
Affiliation(s)
- Seiko Harada
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yo Mabuchi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Haematology, University of Cambridge, Cambridge, UK
| | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Shimojo
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Sadafumi Suzuki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshimi Kawamura
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Kumamoto, Japan
| | - Daisuke Araki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Suyama
- Department of Life Science, Shimane University Faculty of Medicine, Izumo, Shimane, Japan
| | | | - Chihiro Akazawa
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Intractable Disease Research Centre, Juntendo University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yumi Matsuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Department of Life Science, Shimane University Faculty of Medicine, Izumo, Shimane, Japan
| |
Collapse
|
8
|
Alessio N, Stellavato A, Squillaro T, Del Gaudio S, Di Bernardo G, Peluso G, De Rosa M, Schiraldi C, Galderisi U. Hybrid complexes of high and low molecular weight hyaluronan delay in vitro replicative senescence of mesenchymal stromal cells: a pilot study for future therapeutic application. Aging (Albany NY) 2019; 10:1575-1585. [PMID: 30001217 PMCID: PMC6075440 DOI: 10.18632/aging.101493] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/05/2018] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells, a subpopulation of mesenchymal stromal cells (MSCs), are present in the stroma of several tissues. MSC in vitro cultivation for clinical treatments may greatly affect MSC properties. A primary handicap is replicative senescence that impairs MSC functions. Hyaluronan (HA) is present in the extracellular matrix that composes the stem cell niche environment and is under investigation as a key factor for in vitro stem cell growth. We evaluated the effect on MSC cultivation of HA hybrid cooperative complexes (HCC) that are obtained from high (H) and low (L) weight molecules (NAHYCO™). We compared this HCC with H-HA and L-HA. We investigated the effects of these HAs on proliferation, cell cycle, apoptosis, senescence, and differentiation following the addition of the polymer solutions in the culture media at concentrations that did not drastically modify the medium viscosity. Interestingly, 0,16% HCC significantly delayed the senescence compared with the controls. This occurred without alteration of the cell cycle, cytotoxicity, or apoptosis. HCCs also promoted adipogenic and chondrogenic differentiation. Our finding could suggest a potential functional role of HCC above the updated scientific reports of its effects and pave the way to optimization of MSC cultivation for therapeutic application.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonietta Stellavato
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Tiziana Squillaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Stefania Del Gaudio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianfranco Peluso
- Institute of Agri-Environmental Biology and Forestry (IBAF), CNR, Naples, Italy
| | - Mario De Rosa
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Chiara Schiraldi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Umberto Galderisi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19107, USA.,Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy.,Institute of Agri-Environmental Biology and Forestry (IBAF), CNR, Naples, Italy
| |
Collapse
|
9
|
Cheng B, Zhang H, Liu C, Chen X, Chen Y, Sun Y, Leng L, Li Y, Luan P, Li H. Functional Intronic Variant in the Retinoblastoma 1 Gene Underlies Broiler Chicken Adiposity by Altering Nuclear Factor-kB and SRY-Related HMG Box Protein 2 Binding Sites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9727-9737. [PMID: 31398034 DOI: 10.1021/acs.jafc.9b01719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The present study aimed to search for chicken abdominal fat deposition-related polymorphisms within RB1 and to provide functional evidence for significantly associated genetic variants. Association analyses showed that 11 single nucleotide polymorphisms (SNPs) in intron 17 of RB1, were significantly associated with both abdominal fat weight (P < 0.05) and abdominal fat percentage (P < 0.05). Functional analysis revealed that the A allele of g.32828A>G repressed the transcriptional efficiency of RB1 in vitro, through binding nuclear factor-kappa B (NF-KB) and SRY-related HMG box protein 2 (SOX2). Furthermore, RB1 mRNA expression levels in the abdominal fat tissue of individuals with the A/A genotype of g.32828A>G were lower than those of individuals with the G/G genotype. Collectively, we propose that the intronic SNP g.32828A>G of RB1 is an obesity-associated variant that directly affects binding with NF-KB and SOX2, leading to changes in RB1 expression which in turn may influence chicken abdominal fat deposition.
Collapse
Affiliation(s)
- Bohan Cheng
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Hui Zhang
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Chang Liu
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Xi Chen
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Yaofeng Chen
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Yuhang Sun
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Li Leng
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Yumao Li
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Peng Luan
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding , Ministry of Agriculture and Rural Affairs , Harbin 150030 , Heilongjiang , China
- Key Laboratory of Animal Genetics, Breeding and Reproduction , Education Department of Heilongjiang Province , Harbin 150030 , Heilongjiang , China
- College of Animal Science and Technology , Northeast Agricultural University , Harbin 150030 , Heilongjiang , China
| |
Collapse
|
10
|
Alessio N, Squillaro T, Monda V, Peluso G, Monda M, Melone MAB, Galderisi U, Di Bernardo G. Circulating factors present in the sera of naturally skinny people may influence cell commitment and adipocyte differentiation of mesenchymal stromal cells. World J Stem Cells 2019; 11:180-195. [PMID: 30949296 PMCID: PMC6441938 DOI: 10.4252/wjsc.v11.i3.180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/28/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Research on physiopathology of obesity may receive new hints from studies on skinny people (SP). These are individuals who show a poor or null gaining of body weight, in spite of high-calorie intake, by far exceeding the body requirements.
AIM To evaluate how circulating factors present in the SP sera may affect adipogenesis of mesenchymal stromal cells (MSCs).
METHODS We isolated MSCs from bone marrow of healthy donors with both normal body mass index (BMI) and caloric consumption. MSC cultures were primed with sera collected from SP or normal people (NP). Then biomolecular assays were performed to evaluate effect on proliferation, apoptosis, senescence, cell commitment, and differentiation.
RESULTS SP priming affected adipocyte cell commitment and reduced spontaneous adipogenesis. Moreover, an in-depth analysis of exogenous-induced adipocyte differentiation showed striking differences between differentiation in SP-primed samples compared with NP ones. In adipocytes from SP cultures we observed a reduced size of lipid droplets, an increased expression of adipose triglyceride lipase, along with high mitochondria content and ability to produce ATP in starvation condition. These data and the expression of UCP1 protein, indicated that SP pretreatment produced a bias toward brown adipocyte differentiation.
CONCLUSION Our data suggest that sera from SP may promote brown adipogenesis rather that white adipocyte differentiation. This finding could explain why SP present normal body composition in spite of an excess of caloric intake. We hypothesize that some circulating components present in the blood of these individuals may favor brown adipogenesis at expense of white adipocyte production.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Tiziana Squillaro
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Human Physiology and Unit of Dietetic and Sports Medicine Section, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | | | - Marcellino Monda
- Department of Experimental Medicine, Human Physiology and Unit of Dietetic and Sports Medicine Section, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Mariarosa AB Melone
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| |
Collapse
|
11
|
Chen Y, He Y, Wang X, Lu F, Gao J. Adipose‑derived mesenchymal stem cells exhibit tumor tropism and promote tumorsphere formation of breast cancer cells. Oncol Rep 2019; 41:2126-2136. [PMID: 30816504 PMCID: PMC6412463 DOI: 10.3892/or.2019.7018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 01/25/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells reportedly have a marked effect on tumor growth or suppression. However, it remains uncertain whether adipose-derived mesenchymal stem cells (ADSCs) from grafted fat can contribute to breast cancer growth and recurrence. In the present study, interactions between ADSCs and MCF-7 breast cancer cells were evaluated in a Matrigel co-culture system and in an in vivo nude mouse model. Results suggested that MCF-7 cells exerted tumor tropism effects on ADSCs and this may be regulated by chemokines, such as the macrophage inflammatory protein (MIP)-1δ and MIP-3α. Additionally, ADSCs significantly induced tumorsphere formation in vitro and promoted tumorigenicity in vivo. RT-qPCR analysis indicated that tumorsphere formation by MCF-7 cells was associated with the induction of stem-like properties, which was mediated by epithelial-mesenchymal transition. Together, the present findings indicated that ADSCs exhibit tropism and induce tumorsphere formation of MCF-7 cells.
Collapse
Affiliation(s)
- Yanqing Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yunfan He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xuecen Wang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
12
|
Merryweather-Clarke AT, Cook D, Lara BJ, Hua P, Repapi E, Ashley N, Lim SY, Watt SM. Does osteogenic potential of clonal human bone marrow mesenchymal stem/stromal cells correlate with their vascular supportive ability? Stem Cell Res Ther 2018; 9:351. [PMID: 30567594 PMCID: PMC6300038 DOI: 10.1186/s13287-018-1095-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human bone marrow-derived mesenchymal stem/stromal cells (hBM MSCs) have multiple functions, critical for skeletal formation and function. Their functional heterogeneity, however, represents a major challenge for their isolation and in developing potency and release assays to predict their functionality prior to transplantation. Additionally, potency, biomarker profiles and defining mechanisms of action in a particular clinical setting are increasing requirements of Regulatory Agencies for release of hBM MSCs as Advanced Therapy Medicinal Products for cellular therapies. Since the healing of bone fractures depends on the coupling of new blood vessel formation with osteogenesis, we hypothesised that a correlation between the osteogenic and vascular supportive potential of individual hBM MSC-derived CFU-F (colony forming unit-fibroblastoid) clones might exist. METHODS We tested this by assessing the lineage (i.e. adipogenic (A), osteogenic (O) and/or chondrogenic (C)) potential of individual hBM MSC-derived CFU-F clones and determining if their osteogenic (O) potential correlated with their vascular supportive profile in vitro using lineage differentiation assays, endothelial-hBM MSC vascular co-culture assays and transcriptomic (RNAseq) analyses. RESULTS Our results demonstrate that the majority of CFU-F (95%) possessed tri-lineage, bi-lineage or uni-lineage osteogenic capacity, with 64% of the CFU-F exhibiting tri-lineage AOC potential. We found a correlation between the osteogenic and vascular tubule supportive activity of CFU-F clones, with the strength of this association being donor dependent. RNAseq of individual clones defined gene fingerprints relevant to this correlation. CONCLUSIONS This study identified a donor-dependent correlation between osteogenic and vascular supportive potential of hBM MSCs and important gene signatures that support these functions that are relevant to their bone regenerative properties.
Collapse
Affiliation(s)
- Alison T. Merryweather-Clarke
- Stem Cell Research, Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9BQ UK
- Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| | - David Cook
- Stem Cell Research, Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9BQ UK
- Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| | - Barbara Joo Lara
- Stem Cell Research, Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9BQ UK
- Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| | - Peng Hua
- Stem Cell Research, Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9BQ UK
- Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ UK
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| | - Emmanouela Repapi
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| | - Neil Ashley
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| | - Shiang Y. Lim
- Department of Surgery, University of Melbourne, Fitzroy, Victoria 3065 Australia
- O’Brien Institute Department, St. Vincent’s Institute of Medical Research, Fitzroy, Victoria 3065 Australia
| | - Suzanne M. Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9BQ UK
- Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ UK
| |
Collapse
|
13
|
Liu M, Lei H, Dong P, Fu X, Yang Z, Yang Y, Ma J, Liu X, Cao Y, Xiao R. Adipose-Derived Mesenchymal Stem Cells from the Elderly Exhibit Decreased Migration and Differentiation Abilities with Senescent Properties. Cell Transplant 2018; 26:1505-1519. [PMID: 29113467 PMCID: PMC5680952 DOI: 10.1177/0963689717721221] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adipose-derived stem cells (ASCs) can be applied extensively in the clinic because they can be easily isolated and cause less donor-site morbidity; however, their application can be complicated by patient-specific factors, such as age and harvest site. In this study, we systematically evaluated the effects of age on the quantity and quality of human adipose-derived mesenchymal stem cells (hASCs) isolated from excised chest subcutaneous adipose tissue and investigated the underlying molecular mechanism. hASCs were isolated from donors of 3 different age-groups (i.e., child, young adult, and elderly). hASCs are available from individuals across all age-groups and maintain mesenchymal stem cell (MSC) characteristics. However, the increased age of the donors was found to have a significant negative effect on hASCs frequency base on colony-forming unit fibroblasts assay. Moreover, there is a decline in both stromal vascular fraction (SVF) cell yield and the proliferation rate of hASCs with increasing age, although this relationship is not significant. Aging increases cellular senescence, which is manifested as an increase in SA-β-gal-positive cells, increased mitochondrial-specific reactive oxygen species (ROS) production, and the expression of p21 in the elderly. Further, advancing age was found to have a significant negative effect on the adipogenic and osteogenic differentiation potentials of hASCs, particularly at the early and mid-stages of induction, suggesting a slower response to the inducing factors of hASCs from elderly donors. Finally, impaired migration ability was also observed in the elderly group and was determined to be associated with decreased expression of chemokine receptors, such as CXCR4 and CXCR7. Taken together, these results suggest that, while hASCs from different age populations are phenotypically similar, they present major differences at the functional level. When considering potential applications of hASCs in cell-based therapeutic strategies, the negative influence of age on hASC differentiation potential and migration abilities should be taken seriously.
Collapse
Affiliation(s)
- Meichen Liu
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Hua Lei
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ping Dong
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xin Fu
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhigang Yang
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ying Yang
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jiguang Ma
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xia Liu
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yilin Cao
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ran Xiao
- 1 Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
14
|
Pentimalli F, Forte IM, Esposito L, Indovina P, Iannuzzi CA, Alfano L, Costa C, Barone D, Rocco G, Giordano A. RBL2/p130 is a direct AKT target and is required to induce apoptosis upon AKT inhibition in lung cancer and mesothelioma cell lines. Oncogene 2018; 37:3657-3671. [PMID: 29606701 DOI: 10.1038/s41388-018-0214-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 12/30/2017] [Accepted: 02/03/2018] [Indexed: 12/29/2022]
Abstract
The retinoblastoma (RB) protein family includes RB1/p105, RBL1/p107, and RBL2/p130, which are key factors in cell-cycle regulation and stand at the crossroads of multiple pathways dictating cell fate decisions. The role of RB proteins in apoptosis is controversial because they can inhibit or promote apoptosis depending on the context, on the apoptotic stimuli and on their intrinsic status, impacting on the response to antitumoral treatments. Here we identified RBL2/p130 as a direct substrate of the AKT kinase, a key antiapoptotic factor hyperactive in multiple cancer types. We showed that RBL2/p130 and AKT1 physically interact and AKT phosphorylates RBL2/p130 Ser941, located in the pocket domain, but not when this residue is mutated into Ala. We found that pharmacological inhibition of AKT, through the highly selective AKT inhibitor VIII (AKTiVIII), impairs RBL2/p130 Ser941 phosphorylation and increases RBL2/p130 stability, mRNA expression and nuclear levels in both lung cancer and mesothelioma cell lines, mirroring the more extensively studied effects on the p27 cell-cycle inhibitor. Consistently, AKT inhibition reduced cell viability, induced cell accumulation in G0/G1, and triggered apoptosis, which proved to be largely dependent on RBL2/p130 itself, as shown upon RBL2/p130 silencing. AKT inhibition induced RBL2/p130-dependent apoptosis also in HEK-293 cells, in which re-expression of a short hairpin-resistant RBL2/p130 was able to rescue AKTiVIII-induced apoptosis upon RBL2/p130 silencing. Our data also showed that the combination of AKT and cyclin-dependent kinases (CDK) inhibitors, which converge on the re-activation of RBL2/p130 antitumoral potential, could be a promising anticancer strategy.
Collapse
Affiliation(s)
- Francesca Pentimalli
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy.
| | - Iris M Forte
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy
| | - Luca Esposito
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy
| | - Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Scienceand Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Carmelina A Iannuzzi
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy.,Department of Medicine, Surgery and Neuroscience, University of Siena, 53100, Siena, Italy
| | - Luigi Alfano
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy
| | - Caterina Costa
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy
| | - Daniela Barone
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS, "Fondazione G. Pascale", 80131, Napoli, Italy.,Department of Medicine, Surgery and Neuroscience, University of Siena, 53100, Siena, Italy
| | - Gaetano Rocco
- Division of Thoracic Surgery, Department of Thoracic Surgery and Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale"; IRCCS, 80131, Napoli, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Scienceand Technology, Temple University, Philadelphia, PA, 19122, USA. .,Department of Medicine, Surgery and Neuroscience, University of Siena, 53100, Siena, Italy.
| |
Collapse
|
15
|
Münz F, Lopez Perez R, Trinh T, Sisombath S, Weber KJ, Wuchter P, Debus J, Saffrich R, Huber PE, Nicolay NH. Human mesenchymal stem cells lose their functional properties after paclitaxel treatment. Sci Rep 2018; 8:312. [PMID: 29321693 PMCID: PMC5762916 DOI: 10.1038/s41598-017-18862-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an integral part of the bone marrow niche and aid in the protection, regeneration and proliferation of hematopoietic stem cells after exposure to myelotoxic taxane anti-cancer agents, but the influence of taxane compounds on MSCs themselves remains incompletely understood. Here, we show that bone marrow-derived MSCs are highly sensitive even to low concentrations of the prototypical taxane compound paclitaxel. While MSCs remained metabolically viable, they were strongly impaired regarding both their proliferation and their functional capabilities after exposure to paclitaxel. Paclitaxel treatment resulted in reduced cell migration, delays in cellular adhesion and significant dose-dependent inhibition of the stem cells’ characteristic multi-lineage differentiation potential. Cellular morphology and expression of the defining surface markers remained largely unaltered. Paclitaxel only marginally increased apoptosis in MSCs, but strongly induced premature senescence in these stem cells, thereby explaining the preservation of the metabolic activity of functionally inactivated MSCs. The reported sensitivity of MSC function to paclitaxel treatment may help to explain the severe bone marrow toxicities commonly caused by taxane-based anti-cancer treatments.
Collapse
Affiliation(s)
- Franziska Münz
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ramon Lopez Perez
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Thuy Trinh
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Klaus-Josef Weber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Friedrich-Ebert-Str. 107, 68167, Mannheim, Germany
| | - Jürgen Debus
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Friedrich-Ebert-Str. 107, 68167, Mannheim, Germany.,Department of Hematology and Oncology, Heidelberg University Hospital, Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter E Huber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Nils H Nicolay
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Song W, Zhang Y, Wang J, Ma T, Hao L, Wang K. Antagonism of cysteinyl leukotriene receptor 1 (cysLTR1) by montelukast suppresses cell senescence of chondrocytes. Cytokine 2018; 103:83-89. [PMID: 29331588 DOI: 10.1016/j.cyto.2017.12.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 12/22/2022]
Abstract
Aging is closely associated with osteoarthritis (OA). Although its underlying mechanisms remain unknown, cellular senescence in chondrocytes has become an important therapeutic target for the treatment of OA. Cysteinyl leukotriene receptors (cysLTRs) mediate the pathobiological function of cysteinyl leukotrienes (cysLTs). However, the roles of cysLTRs in the pathogenesis of OA have not been reported before. In the current study, we found that cysLTR1 but not cysLTR2 is expressed in human primary chondrocytes. In addition, stimulation with tumor necrosis factor α (TNF-α) resulted in a significant increase in the expression of cysLTR1. Interestingly, montelukast, a specific cysLTR1 antagonist, attenuated TNF-α-induced up-regulation of the activity of senescence-associated β-galactosidase (SA-β-Gal). In addition, TNF-α led to cell cycle arrest at the G0/G1 phase, which was prevented by treatment with montelukast. Notably, montelukast reduced expression of the senescence markers p53, p21 and PAI-1. In addition, montelukast ameliorated TNF-α-induced K382 acetylation of p53 by promoting the expression of SIRT1. Silencing of SIRT1 using SIRT1 siRNA broke the inhibitory effects of montelukast on K382 acetylation of p53. Importantly, silencing of cysLTR1 reversed the reduction of SIRT1 expression as well as the K382 acetylation of p53. Our findings strongly implicate that cysLTR1 has the capacity to regulate cellular senescence in chondrocytes. It is suggested that montelukast may be a potential therapeutic agent for chondro-protective therapy.
Collapse
Affiliation(s)
- Wei Song
- Medical School, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yumin Zhang
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Jun Wang
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Tao Ma
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Linjie Hao
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Kunzheng Wang
- First Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
17
|
Rühle A, Xia O, Perez RL, Trinh T, Richter W, Sarnowska A, Wuchter P, Debus J, Saffrich R, Huber PE, Nicolay NH. The Radiation Resistance of Human Multipotent Mesenchymal Stromal Cells Is Independent of Their Tissue of Origin. Int J Radiat Oncol Biol Phys 2018; 100:1259-1269. [PMID: 29452769 DOI: 10.1016/j.ijrobp.2018.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/10/2017] [Accepted: 01/03/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE Human mesenchymal stromal cells (MSCs) may aid the regeneration of ionizing radiation (IR)-induced tissue damage. They can be harvested from different tissues for clinical purposes; however, the role of the tissue source on the radiation response of human MSCs remains unknown. METHODS AND MATERIALS Human MSCs were isolated from adipose tissue, bone marrow, and umbilical cord, and cellular survival, proliferation, and apoptosis were measured after irradiation. The influence of IR on the defining functions of MSCs was assessed, and cell morphology, surface marker expression, and the differentiation potential were examined. Western blot analyses were performed to assess the activation of DNA damage signaling and repair pathways. RESULTS MSCs from adipose tissue, bone marrow, and umbilical cord exhibited a relative radioresistance independent of their tissue of origin. Defining properties including cellular adhesion and surface marker expression were preserved, and irradiated MSCs maintained their potential for multilineage differentiation irrespective of their tissue source. Analysis of activated DNA damage recognition and repair pathways demonstrated an efficient repair of IR-induced DNA double-strand breaks in MSCs from different tissues, thereby influencing the induction of apoptosis. CONCLUSIONS These data show for the first time that MSCs are resistant to IR and largely preserve their defining functions after irradiation irrespective of their tissue of origin. Efficient repair of IR-induced DNA double-strand breaks and consecutive reduction of apoptosis induction may contribute to the tissue-independent radiation resistance of MSCs.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Xia
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna Sarnowska
- Translative Platform for Regenerative Medicine, Mossakowski Medical Research Center, Polish Academy of Sciences, Warsaw, Poland
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Donor Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Donor Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; Department of Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
18
|
Yu J, Shi J, Zhang Y, Zhang Y, Huang Y, Chen Z, Yang J. The replicative senescent mesenchymal stem / stromal cells defect in DNA damage response and anti-oxidative capacity. Int J Med Sci 2018; 15:771-781. [PMID: 30008586 PMCID: PMC6036081 DOI: 10.7150/ijms.24635] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 04/27/2018] [Indexed: 01/11/2023] Open
Abstract
Replicative senescence and potential malignant transformation are great limitations in the clinical application of bone marrow-derived mesenchymal stem / stromal cells (MSCs). An abnormal DNA damage response may result in genomic instability, which is an integral component of aging and tumorigenesis. However, the effect of aging on the DNA damage response in MSCs is currently unknown. In the present study, we evaluated the DNA damage response induced by oxidative stress and DNA double-strand breaks in human bone marrow-derived MSCs. After long-term cell culture, replicative senescent MSCs (sMSCs) were characterized by a poor proliferation rate, high senescence-associated β-galactosidase activity, and enhanced expression of P53 and P16. Features of the DNA damage response in these sMSCs were then compared with those from early-passage MSCs. The sMSCs were more sensitive to hydrogen peroxide and bleomycin treatment with respect to cell viability and apoptosis induction. Combined with the comet assay, γH2AX foci characterization and reactive oxygen species detection were used to demonstrate that the antioxidant and DNA repair ability of sMSCs are attenuated. This result could be explained, at least in part, by the downregulation of anti-oxidation and DNA repair genes, including Cu/Zn-SOD, GPX, CAT, OGG1, XRCC1, Ku70, BRCA2 and XRCC4. In conclusion, MSCs aging is associated with a reduction in the DNA repair and anti-oxidative capacity.
Collapse
Affiliation(s)
- Jin Yu
- Department of Cell Biology, Army Medical University (The Third Military Medical University), Chongqing 400038, China
| | - Jiazhong Shi
- Department of Cell Biology, Army Medical University (The Third Military Medical University), Chongqing 400038, China
| | - Yue Zhang
- Department of Cell Biology, Army Medical University (The Third Military Medical University), Chongqing 400038, China.,Department of Pathology, The 451th hospital of PLA, Xi'an 710000, China
| | - Yi Zhang
- Department of Cell Biology, Army Medical University (The Third Military Medical University), Chongqing 400038, China
| | - Yaqin Huang
- Department of Cell Biology, Army Medical University (The Third Military Medical University), Chongqing 400038, China
| | - Zhiwen Chen
- Urology Institute of PLA, Southwest Hospital, Army Medical University (The Third Military Medical University), Chongqing 400038, China
| | - Jin Yang
- Department of Cell Biology, Army Medical University (The Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
19
|
Kundrotas G, Gasperskaja E, Slapsyte G, Gudleviciene Z, Krasko J, Stumbryte A, Liudkeviciene R. Identity, proliferation capacity, genomic stability and novel senescence markers of mesenchymal stem cells isolated from low volume of human bone marrow. Oncotarget 2017; 7:10788-802. [PMID: 26910916 PMCID: PMC4905439 DOI: 10.18632/oncotarget.7456] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/05/2016] [Indexed: 12/16/2022] Open
Abstract
Human bone marrow mesenchymal stem cells (hBM-MSCs) hold promise for treating incurable diseases and repairing of damaged tissues. However, hBM-MSCs face the disadvantages of painful invasive isolation and limited cell numbers. In this study we assessed characteristics of MSCs isolated from residual human bone marrow transplantation material and expanded to clinically relevant numbers at passages 3-4 and 6-7. Results indicated that early passage hBM-MSCs are genomically stable and retain identity and high proliferation capacity. Despite the chromosomal stability, the cells became senescent at late passages, paralleling the slower proliferation, altered morphology and immunophenotype. By qRT-PCR array profiling, we revealed 13 genes and 33 miRNAs significantly differentially expressed in late passage cells, among which 8 genes and 30 miRNAs emerged as potential novel biomarkers of hBM-MSC aging. Functional analysis of genes with altered expression showed strong association with biological processes causing cellular senescence. Altogether, this study revives hBM as convenient source for cellular therapy. Potential novel markers provide new details for better understanding the hBM-MSC senescence mechanisms, contributing to basic science, facilitating the development of cellular therapy quality control, and providing new clues for human disease processes since senescence phenotype of the hematological patient hBM-MSCs only very recently has been revealed.
Collapse
Affiliation(s)
- Gabrielis Kundrotas
- Department of Botany and Genetics, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania.,Biobank, National Cancer Institute, Vilnius, Lithuania
| | - Evelina Gasperskaja
- Department of Botany and Genetics, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania
| | - Grazina Slapsyte
- Department of Botany and Genetics, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania
| | | | - Jan Krasko
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| | | | | |
Collapse
|
20
|
Gehrke T, Scherzad A, Hackenberg S, Ickrath P, Schendzielorz P, Hagen R, Kleinsasser N. Long-term changes in the properties of skin-derived fibroblasts following irradiation of the head and neck. Oncol Lett 2017; 14:3780-3786. [PMID: 28927147 DOI: 10.3892/ol.2017.6593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/03/2017] [Indexed: 01/03/2023] Open
Abstract
The tumor stroma performs an important role in carcinogenesis. It predominantly consists of fibroblasts and the connective tissue produced by them, and undergoes a multitude of interactions with the surrounding cancer cells. Since irradiation is part of the majority of therapeutic strategies for head and neck squamous cell carcinoma, more information regarding the effects of a previous irradiation on the tumor stroma is desirable. In the present study, fibroblasts were cultivated from human non-irradiated and pre-irradiated skin of the neck for 48 h. Subsequently, analyses of cell viability, apoptosis, necrosis and motility were conducted via MTT assay, Annexin V/propidium iodide staining, electronic cell counting for 4 consecutive days, and scratch assay. Pre-irradiated fibroblasts exhibited a significantly slower growth rate as well as increased rates of apoptosis and necrosis. They also exhibited significantly decreased motility compared with non-irradiated fibroblasts. These results indicated the long-term effects of irradiation on fibroblasts, which may affect cancer recurrence in the irradiated region via the tumor stroma. More information, such as that regarding the secretory capacities of pre-irradiated fibroblasts, is required to evaluate the possible therapeutic implications of these findings.
Collapse
Affiliation(s)
- Thomas Gehrke
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Agmal Scherzad
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Pascal Ickrath
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Philipp Schendzielorz
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Rudolf Hagen
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| | - Norbert Kleinsasser
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Würzburg, D-97080 Würzburg, Germany
| |
Collapse
|
21
|
Chen L, Ran Q, Xiang Y, Xiang L, Chen L, Li F, Wu J, Wu C, Li Z. Co-Activation of PKC-δ by CRIF1 Modulates Oxidative Stress in Bone Marrow Multipotent Mesenchymal Stromal Cells after Irradiation by Phosphorylating NRF2 Ser40. Theranostics 2017; 7:2634-2648. [PMID: 28819452 PMCID: PMC5558558 DOI: 10.7150/thno.17853] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 04/19/2017] [Indexed: 12/26/2022] Open
Abstract
The high mortality associated with pancytopenia and multi-organ failure resulting from hematopoietic disorders of acute radiation syndrome (h-ARS) creates an urgent need for developing more effective treatment strategies. Here, we showed that bone marrow multipotent mesenchymal stromal cells (BMMSCs) effectively regulate oxidative stress following radiative injury, which might be on account of irradiation-induced elevation of protein levels of CR6-interacting factor 1(CRIF1) and nuclear factor E2-related factor 2(NRF2). Crif1-knockdown BMMSCs presented increased oxidative stress and apoptosis after irradiation, which were partially due to a suppressed antioxidant response mediated by decreased NRF2 nuclear translocation. Co-immunoprecipitation (Co-IP) experiments indicated that CRIF1 interacted with protein kinase C-δ (PKC-δ). NRF2 Ser40 phosphorylation was inhibited in Crif1-deficient BMMSCs even in the presence of three kinds of PKC agonists, suggesting that CRIF1 might co-activate PKC-δ to phosphorylate NRF2 Ser40. After radiative injury, the supporting effect of BMMSCs for the colony forming ability of HSCs in vitro was reduced, and the deficiency of CRIF1 aggravated such damage. Thus, CRIF1 plays an essential role in PKC-δ/NRF2 pathway modulation to alleviate oxidative stress in BMMSCs after irradiative injury, and at some level it may maintain the HSCs-supporting effect of BMMSCs after radiative injuries.
Collapse
|
22
|
Liu J, Peng L, Huang W, Li Z, Pan J, Sang L, Lu S, Zhang J, Li W, Luo Y. Balancing Between Aging and Cancer: Molecular Genetics Meets Traditional Chinese Medicine. J Cell Biochem 2017; 118:2581-2586. [DOI: 10.1002/jcb.25898] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/18/2017] [Indexed: 01/23/2023]
Affiliation(s)
- Jing Liu
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
| | - Lei Peng
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
| | - Wenhui Huang
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
| | - Zhiming Li
- Institute of Medicinal Plants; Yunnan Academy of Agricultural Sciences; Kunming 650200 China
| | - Jun Pan
- Institute of Medicinal Plants; Yunnan Academy of Agricultural Sciences; Kunming 650200 China
| | - Lei Sang
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
| | - Siqian Lu
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
| | - Jihong Zhang
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
| | - Wanyi Li
- Institute of Medicinal Plants; Yunnan Academy of Agricultural Sciences; Kunming 650200 China
| | - Ying Luo
- Lab of Molecular Genetics of Aging and Tumor; Faculty of Medicine; Kunming University of Science and Technology; Chenggong County, Kunming Yunnan Province 650500 China
- Yunnan Provincial Institute of Digestive Disease; Kunming; Yunnan Province 650011 China
| |
Collapse
|
23
|
Alessio N, Esposito G, Galano G, De Rosa R, Anello P, Peluso G, Tabocchini MA, Galderisi U. Irradiation of Mesenchymal Stromal Cells With Low and High Doses of Alpha Particles Induces Senescence and/or Apoptosis. J Cell Biochem 2017; 118:2993-3002. [PMID: 28252222 DOI: 10.1002/jcb.25961] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/28/2017] [Indexed: 12/25/2022]
Abstract
The use of high-linear energy transfer charged particles is gaining attention as a medical tool because of the emission of radiations with an efficient cell-killing ability. Considerable interest has developed in the use of targeted alpha-particle therapy for the treatment of micrometastases. Moreover, the use of helium beams is gaining momentum, especially for treating pediatric tumors. We analyzed the effects of alpha particles on bone marrow mesenchymal stromal cells (MSCs), which have a subpopulation of stem cells capable of generating adipocytes, chondrocytes, and osteocytes. Further, these cells contribute toward maintenance of homeostasis in the body. MSCs were irradiated with low and high doses of alpha particles or X-rays and a comparative biological analysis was performed. At a low dose (40 mGy), alpha particles exhibited a limited negative effect on the biology of MSCs compared with X-rays. No significant perturbation of cell cycle was observed, and a minimal increase in apoptosis or senescence was detected. Self-renewal was preserved as revealed by the CFU assay. On the contrary, with 2000 mGy alpha particles, we observed adverse effects on the vitality, functionality, and stemness of MSCs. These results are the consequence of different proportion of cells targeted by alpha particles or X-rays and the quality of induced DNA damage. The present study suggests that radiotherapy with alpha particles may spare healthy stem cells more efficaciously than X-ray treatments, an observation that should be taken into consideration by physicians while planning irradiation of tumor areas close to stem cell niches, such as bone marrow. J. Cell. Biochem. 118: 2993-3002, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Campania University "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Esposito
- Technology and Health Department, National Institute of Health, Rome, Italy.,National Institute of Nuclear Physics, Section Roma 1, Rome, Italy
| | - Giovanni Galano
- PSI Napoli Est - Laboratory UO, ASL Napoli 1 Centro, Naples, Italy
| | - Roberto De Rosa
- PSI Napoli Est - Radiology UO, ASL Napoli 1 Centro, Naples, Italy
| | - Pasquale Anello
- Technology and Health Department, National Institute of Health, Rome, Italy
| | - Gianfranco Peluso
- Institute of Agro-Environmental Biology and Forestry (IBAF), CNR, Naples, Italy
| | - Maria Antonella Tabocchini
- Technology and Health Department, National Institute of Health, Rome, Italy.,National Institute of Nuclear Physics, Section Roma 1, Rome, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Campania University "Luigi Vanvitelli", Naples, Italy.,Institute of Agro-Environmental Biology and Forestry (IBAF), CNR, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Molecular connections of obesity and aging: a focus on adipose protein 53 and retinoblastoma protein. Biogerontology 2017; 18:321-332. [PMID: 28357524 DOI: 10.1007/s10522-017-9698-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/27/2017] [Indexed: 12/13/2022]
Abstract
Obesity is an induced health problem that human beings have been facing with non-optimal treatment so far. Humans are on average getting fatter with age, and obesity and aging interact each other to shorten lifetime and decrease life quality. Obesity also causes several aging related-disorders such as cancer, strokes, cardiovascular disease, high blood pressure and type 2 diabetes. So, the molecular connections between aging and obesity are promising targets for bio-medical researches and innovative therapies of many health problems. In this review, we discuss the findings of adipose p53 and Rb-two central molecular linkages between aging and obesity-on lipid metabolism and obesity.
Collapse
|
25
|
Li Y, Wu Q, Wang Y, Li L, Bu H, Bao J. Senescence of mesenchymal stem cells (Review). Int J Mol Med 2017; 39:775-782. [PMID: 28290609 DOI: 10.3892/ijmm.2017.2912] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 01/13/2017] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in cell-based therapy for various diseases, due to their immunomodulatory and inflammatory effects. However, the function of MSCs is known to decline with age, a process that is called senescence. To date, the process of MSC senescence remains unknown as in-depth understanding of the mechanisms involved in cellular senescence is lacking. First, senescent MSCs are so heterogeneous that not all of them express the same phenotypic markers. In addition, the genes and signaling pathways which regulate this process in MSCs are still unknown. Thus, an understanding of the molecular processes controlling MSC senescence is crucial to determining the drivers and effectors of age-associated MSC dysfunction. Moreover, the proper use of MSCs for clinical application requires a general understanding of the MSC aging process. Furthermore, such knowledge is essential for the development of therapeutic interventions that can slow or reverse age-related degenerative changes to enhance repair processes and maintain healthy function in aging tissues. To further clarify the properties of senescent cells, as well as to present significant findings from studies on the mechanisms of cellular aging, we summarize these biological features in the senescence of MSCs in this scenario. This review summarizes recent advances in our understanding of the markers and differentiation potential indicating MSC senescence, as well as factors affecting MSC senescence with particular emphasis on the roles of oxidative stress, intrinsic changes in telomere shortening, histone deacetylase and DNA methyltransferase, genes and signaling pathways and immunological properties.
Collapse
Affiliation(s)
- Yi Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qiong Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yujia Wang
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Bu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ji Bao
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
26
|
Musto P, Calarco A, Pannico M, La Manna P, Margarucci S, Tafuri A, Peluso G. Hyperspectral Raman imaging of human prostatic cells: An attempt to differentiate normal and malignant cell lines by univariate and multivariate data analysis. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2017; 173:476-488. [PMID: 27718451 DOI: 10.1016/j.saa.2016.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 08/29/2016] [Accepted: 09/19/2016] [Indexed: 06/06/2023]
Abstract
Hyperspectral Raman images of human prostatic cells have been collected and analysed with several approaches to reveal differences among normal and tumor cell lines. The objective of the study was to test the potential of different chemometric methods in providing diagnostic responses. We focused our analysis on the ν(CH) region (2800-3100cm-1) owing to its optimal Signal-to-Noise ratio and because the main differences between the spectra of the two cell lines were observed in this frequency range. Multivariate analysis identified two principal components, which were positively recognized as due to the protein and the lipid fractions, respectively. The tumor cells exhibited a modified distribution of the cytoplasmatic lipid fraction (mainly localized alongside the cell boundary) which may result very useful for a preliminary screening. Principal Component analysis was found to provide high contrast and to be well suited for image-processing purposes. Self-Modelling Curve Resolution made available meaningful spectra and relative-concentration values; it revealed a 97% increase of the lipid fraction in the tumor cell with respect to the control. Finally, a univariate approach confirmed significant and reproducible differences between normal and tumor cells.
Collapse
Affiliation(s)
- P Musto
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, via Campi Flegrei, 34, Olivetti Buildings, 80078 Pozzuoli, NA, Italy.
| | - A Calarco
- Institute of Biosciences and Bio Resources, National Research Council of Italy, via P. Castellino 111, 80131 Naples, NA, Italy
| | - M Pannico
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, via Campi Flegrei, 34, Olivetti Buildings, 80078 Pozzuoli, NA, Italy
| | - P La Manna
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, via Campi Flegrei, 34, Olivetti Buildings, 80078 Pozzuoli, NA, Italy
| | - S Margarucci
- Institute of Biosciences and Bio Resources, National Research Council of Italy, via P. Castellino 111, 80131 Naples, NA, Italy
| | - A Tafuri
- Hematology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa, n. 1035, 00189 Rome, Italy
| | - G Peluso
- Institute of Biosciences and Bio Resources, National Research Council of Italy, via P. Castellino 111, 80131 Naples, NA, Italy
| |
Collapse
|
27
|
Squillaro T, Antonucci I, Alessio N, Esposito A, Cipollaro M, Melone MAB, Peluso G, Stuppia L, Galderisi U. Impact of lysosomal storage disorders on biology of mesenchymal stem cells: Evidences from in vitro silencing of glucocerebrosidase (GBA) and alpha-galactosidase A (GLA) enzymes. J Cell Physiol 2017; 232:3454-3467. [PMID: 28098348 DOI: 10.1002/jcp.25807] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 12/27/2022]
Abstract
Lysosomal storage disorders (LDS) comprise a group of rare multisystemic diseases resulting from inherited gene mutations that impair lysosomal homeostasis. The most common LSDs, Gaucher disease (GD), and Fabry disease (FD) are caused by deficiencies in the lysosomal glucocerebrosidase (GBA) and alpha-galactosidase A (GLA) enzymes, respectively. Given the systemic nature of enzyme deficiency, we hypothesized that the stem cell compartment of GD and FD patients might be also affected. Among stem cells, mesenchymal stem cells (MSCs) are a commonly investigated population given their role in hematopoiesis and the homeostatic maintenance of many organs and tissues. Since the impairment of MSC functions could pose profound consequences on body physiology, we evaluated whether GBA and GLA silencing could affect the biology of MSCs isolated from bone marrow and amniotic fluid. Those cell populations were chosen given the former's key role in organ physiology and the latter's intriguing potential as an alternative stem cell model for human genetic disease. Our results revealed that GBA and GLA deficiencies prompted cell cycle arrest along with the impairment of autophagic flux and an increase of apoptotic and senescent cell percentages. Moreover, an increase in ataxia-telangiectasia-mutated staining 1 hr after oxidative stress induction and a return to basal level at 48 hr, along with persistent gamma-H2AX staining, indicated that MSCs properly activated DNA repair signaling, though some damages remained unrepaired. Our data therefore suggest that MSCs with reduced GBA or GLA activity are prone to apoptosis and senescence due to impaired autophagy and DNA repair capacity.
Collapse
Affiliation(s)
- Tiziana Squillaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy.,Institute of Bioscience and Bioresources, National Research Council, Naples, Italy
| | - Ivana Antonucci
- Laboratory of Molecular Genetics, Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Anna Esposito
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mariarosa Anna Beatrice Melone
- Department of Medical, Surgical, Neurological, Metabolic Sciences, and Aging; Division of Neurology and InterUniversity Center for Research in Neurosciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianfranco Peluso
- Institute of Bioscience and Bioresources, National Research Council, Naples, Italy
| | - Liborio Stuppia
- Laboratory of Molecular Genetics, Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, G. d'Annunzio University, Chieti-Pescara, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
28
|
Yang R, Wu L, Chen J, Chen W, Zhang L, Zhang L, You R, Yin L, Li CH, Guan YQ. Effects of Differentiation and Antisenescence from BMSCs to Hepatocy-Like Cells of the PAAm-IGF-1/TNF-α Biomaterial. ACS APPLIED MATERIALS & INTERFACES 2016; 8:26638-26647. [PMID: 27668443 DOI: 10.1021/acsami.6b10377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Aiming at the cells' differentiation phenomenon and senescence problem in liver tissue engineering, this work is designed to synthesize three different chargeable polymers (polypropylene acid (PAAc), polyethylene glycol (PEG), and polypropylene amine (PAAm)) coimmobilized by the insulin-like growth factor 1 (IGF-1) and tumor necrosis factor-α (TNF-α). We explore the hepatocyte differentiation effect and the antisenecence effect of PSt-PAAm-IGF-1/TNF-α biomaterial which was selected from the three different chargeable polymers in bone marrow mesenchymal stem cells (BMSCs). Our work will establish a model for studying the biochemical molecular regulation mechanism and signal transduction pathway of cell senescence in liver tissue engineering, which provide a molecular basis for developing biomaterials for liver tissue engineering.
Collapse
Affiliation(s)
- Runcai Yang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Lifang Wu
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Jiehong Chen
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Wuya Chen
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Lin Zhang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Li Zhang
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Rong You
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University , Guangzhou 510631, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University , Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University , Guangzhou 510631, China
| |
Collapse
|
29
|
Özcan S, Alessio N, Acar MB, Toprak G, Gönen ZB, Peluso G, Galderisi U. Myeloma cells can corrupt senescent mesenchymal stromal cells and impair their anti-tumor activity. Oncotarget 2016; 6:39482-92. [PMID: 26498687 PMCID: PMC4741840 DOI: 10.18632/oncotarget.5430] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/05/2015] [Indexed: 12/23/2022] Open
Abstract
Senescent cells secrete several molecules that help to prevent the progression of cancer. However, cancer cells can also misuse these secreted elements to survive and grow. Since the molecular and functional bases of these different elements remain poorly understood, we analyzed the effect of senescent mesenchymal stromal cell (MSC) secretome on the biology of ARH-77 myeloma cells. In addition to differentiating in mesodermal derivatives, MSCs have sustained interest among researchers by supporting hematopoiesis, contributing to tissue homeostasis, and modulating inflammatory response, all activities accomplished primarily by the secretion of cytokines and growth factors. Moreover, senescence profoundly affects the composition of MSC secretome. In this study, we induced MSC senescence by oxidative stress, DNA damage, and replicative exhaustion. While the first two are considered to induce acute senescence, extensive proliferation triggers replicative (i.e., chronic) senescence. We cultivated cancer cells in the presence of acute and chronic senescent MSC-conditioned media and evaluated their proliferation, DNA damage, apoptosis, and senescence. Our findings revealed that senescent secretomes induced apoptosis or senescence, if not both, to different extents. This anti-tumor activity became heavily impaired when secretomes were collected from senescent cells previously in contact (i.e., primed) with cancer cells. Our analysis of senescent MSC secretomes with LC-MS/MS followed by Gene Ontology classification further indicated that priming with cancer profoundly affected secretome composition by abrogating the production of pro-senescent and apoptotic factors. We thus showed for the first time that compared with cancer-primed MSCs, naïve senescent MSCs can exert different effects on tumor progression.
Collapse
Affiliation(s)
- Servet Özcan
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey.,Department of Biology, Faculty of Sciences, Erciyes University, Kayseri, Turkey
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Mustafa Burak Acar
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey.,Graduate School of Natural and Applied Sciences, Erciyes University, Kayseri, Turkey
| | - Güler Toprak
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | | | | | - Umberto Galderisi
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey.,Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
30
|
Senescence in Human Mesenchymal Stem Cells: Functional Changes and Implications in Stem Cell-Based Therapy. Int J Mol Sci 2016; 17:ijms17071164. [PMID: 27447618 PMCID: PMC4964536 DOI: 10.3390/ijms17071164] [Citation(s) in RCA: 346] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/04/2016] [Accepted: 07/14/2016] [Indexed: 12/19/2022] Open
Abstract
Regenerative medicine is extensively interested in developing cell therapies using mesenchymal stem cells (MSCs), with applications to several aging-associated diseases. For successful therapies, a substantial number of cells are needed, requiring extensive ex vivo cell expansion. However, MSC proliferation is limited and it is quite likely that long-term culture evokes continuous changes in MSCs. Therefore, a substantial proportion of cells may undergo senescence. In the present review, we will first present the phenotypic characterization of senescent human MSCs (hMSCs) and their possible consequent functional alterations. The accumulation of oxidative stress and dysregulation of key differentiation regulatory factors determine decreased differentiation potential of senescent hMSCs. Senescent hMSCs also show a marked impairment in their migratory and homing ability. Finally, many factors present in the secretome of senescent hMSCs are able to exacerbate the inflammatory response at a systemic level, decreasing the immune modulation activity of hMSCs and promoting either proliferation or migration of cancer cells. Considering the deleterious effects that these changes could evoke, it would appear of primary importance to monitor the occurrence of senescent phenotype in clinically expanded hMSCs and to evaluate possible ways to prevent in vitro MSC senescence. An updated critical presentation of the possible strategies for in vitro senescence monitoring and prevention constitutes the second part of this review. Understanding the mechanisms that drive toward hMSC growth arrest and evaluating how to counteract these for preserving a functional stem cell pool is of fundamental importance for the development of efficient cell-based therapeutic approaches.
Collapse
|
31
|
Alessio N, Capasso S, Di Bernardo G, Cappabianca S, Casale F, Calarco A, Cipollaro M, Peluso G, Galderisi U. Mesenchymal stromal cells having inactivated RB1 survive following low irradiation and accumulate damaged DNA: Hints for side effects following radiotherapy. Cell Cycle 2016; 16:251-258. [PMID: 27124644 DOI: 10.1080/15384101.2016.1175798] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Following radiotherapy, bone sarcomas account for a significant percentage of recurring tumors. This risk is further increased in patients with hereditary retinoblastoma that undergo radiotherapy. We analyzed the effect of low and medium dose radiation on mesenchymal stromal cells (MSCs) with inactivated RB1 gene to gain insights on the molecular mechanisms that can induce second malignant neoplasm in cancer survivors. MSC cultures contain subpopulations of mesenchymal stem cells and committed progenitors that can differentiate into mesodermal derivatives: adipocytes, chondrocytes, and osteocytes. These stem cells and committed osteoblast precursors are the cell of origin in osteosarcoma, and RB1 gene mutations have a strong role in its pathogenesis. Following 40 and 2000 mGy X-ray exposure, MSCs with inactivated RB1 do not proliferate and accumulate high levels of unrepaired DNA as detected by persistence of gamma-H2AX foci. In samples with inactivated RB1 the radiation treatment did not increase apoptosis, necrosis or senescence versus untreated cells. Following radiation, CFU analysis showed a discrete number of cells with clonogenic capacity in cultures with silenced RB1. We extended our analysis to the other members of retinoblastoma gene family: RB2/P130 and P107. Also in the MSCs with silenced RB2/P130 and P107 we detected the presence of cells with unrepaired DNA following X-ray irradiation. Cells with unrepaired DNA may represent a reservoir of cells that may undergo neoplastic transformation. Our study suggests that, following radiotherapy, cancer patients with mutations of retinoblastoma genes may be under strict controls to evaluate onset of secondary neoplasms following radiotherapy.
Collapse
Affiliation(s)
- Nicola Alessio
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy
| | - Stefania Capasso
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy
| | - Giovanni Di Bernardo
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy
| | - Salvatore Cappabianca
- b Department "F. Magrassi - A. Lanzara" Second University of Naples , Naples , Italy
| | - Fiorina Casale
- c Dipartimento della Donna , del Bambino e di Chirurgia Generale e Specialistica, Second University of Naples , Naples , Italy
| | - Anna Calarco
- d Institute of Bioscience and Bioresources, CNR , Naples , Italy
| | - Marilena Cipollaro
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy
| | - Gianfranco Peluso
- b Department "F. Magrassi - A. Lanzara" Second University of Naples , Naples , Italy
| | - Umberto Galderisi
- a Department of Experimental Medicine , Biotechnology and Molecular Biology Section, Second University of Naples , Naples , Italy
| |
Collapse
|
32
|
Squillaro T, Severino V, Alessio N, Farina A, Di Bernardo G, Cipollaro M, Peluso G, Chambery A, Galderisi U. De-regulated expression of the BRG1 chromatin remodeling factor in bone marrow mesenchymal stromal cells induces senescence associated with the silencing of NANOG and changes in the levels of chromatin proteins. Cell Cycle 2016; 14:1315-26. [PMID: 25724006 DOI: 10.4161/15384101.2014.995053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Stem cells have a peculiar chromatin architecture that contributes to their unique properties, including uncommitted status, multi/pluripotency and self-renewal. We analyzed the effect of the de-regulation of the SWI/SNF chromatin remodeling complex in mesenchymal stromal cells (MSC) through the silencing and up-regulation of BRG1, which is the ATPase subunit of the complex. The altered expression of BRG1 promoted the senescence of MSC with suppression of the NANOG transcription, which is part of the transcriptional circuitry governing stem cell functions. To gain insight on the way NANOG was silenced, we evaluated how the de-regulated BRG1 expression affect the binding of activators and repressors on the NANOG promoter. We found 4 E2F binding motifs on NANOG promoter, which can be occupied by RB1 and RB2/P130. These are members of the retinoblastoma gene family. In MSC with a silenced BRG1, the relative binding of the 2 retinoblastoma proteins increased, and this was associated with the recruitment of DNMT1. This induced the methylation of CpG on the NANOG promoter. Opposingly, when a high level of BRG1 was present, the same E2F binding motifs were docking sites for BRG1, which induced chromatin compaction without CpG methylation but with increased histone deacetylation, associated with the presence of HDAC1 on E2F binding sites. Besides the sharp regulation of the NANOG expression, we evidenced, through proteomic analysis, that the de-regulation of the SWI/SNF function affected the expression of histones and other nuclear proteins involved in "nuclear architecture," suggesting that BRG1 may act as global regulator of gene expression.
Collapse
Affiliation(s)
- Tiziana Squillaro
- a Department of Experimental Medicine; Biotechnology and Molecular Biology Section ; Second University of Naples ; Naples , Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Nicolay NH, Perez RL, Saffrich R, Huber PE. Radio-resistant mesenchymal stem cells: mechanisms of resistance and potential implications for the clinic. Oncotarget 2015; 6:19366-80. [PMID: 26203772 PMCID: PMC4637291 DOI: 10.18632/oncotarget.4358] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 05/30/2015] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) comprise a heterogeneous population of multipotent stromal cells and can be isolated from various tissues and organs. Due to their regenerative potential, they have been subject to intense research efforts, and they may provide an efficient means for treating radiation-induced tissue damage. MSCs are relatively resistant to ionizing radiation and retain their stem cell characteristics even after high radiation doses. The underlying mechanisms for the observed MSC radioresistance have been extensively studied and may involve efficient DNA damage recognition, double strand break repair and evasion of apoptosis. Here, we present a concise review of the published scientific data on the radiobiological features of MSCs. The involvement of different DNA damage recognition and repair pathways in the creation of a radioresistant MSC phenotype is outlined, and the roles of apoptosis, senescence and autophagy regarding the reported radioresistance are summarized. Finally, potential influences of the radioresistant MSCs for the clinic are discussed with respect to the repair and radioprotection of irradiated tissues.
Collapse
Affiliation(s)
- Nils H. Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Ramon Lopez Perez
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter E. Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| |
Collapse
|
34
|
Lin ZB, Qian B, Yang YZ, Zhou K, Sun J, Mo XM, Wu KH. Isolation, Characterization and Cardiac Differentiation of Human Thymus Tissue Derived Mesenchymal Stromal Cells. J Cell Biochem 2015; 116:1205-12. [PMID: 25535722 DOI: 10.1002/jcb.25072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 12/18/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Ze Bang Lin
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Bo Qian
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Yu Zhong Yang
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Kai Zhou
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Jian Sun
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Xu Ming Mo
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| | - Kai Hong Wu
- Department of Cardiothoracic Surgery; Nanjing Children‘s Hospital; Nanjing Medical University; Nanjing China
| |
Collapse
|
35
|
Cao HJ, Zheng LZ, Wang N, Wang LY, Li Y, Li D, Lai YX, Wang XL, Qin L. Src blockage by siRNA inhibits VEGF-induced vascular hyperpemeability and osteoclast activity - an in vitro mechanism study for preventing destructive repair of osteonecrosis. Bone 2015; 74:58-68. [PMID: 25554601 DOI: 10.1016/j.bone.2014.12.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 12/08/2014] [Accepted: 12/23/2014] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Destructive repair is the pathological feature of ONFH characterized with the elevated vascular permeability and persistent bone resorption, which is associated with higher VEGF expression, activated c-Src, and vascular leakage. Activated c-Src also participates in mediating endothelial permeability and osteoclasts activity. However, the molecular mechanism of the VEGF and c-Src contributing to the destructive repair process remains unknown. The purpose of this study is to delineate the role of VEGF and c-Src in triggering destructive repair of osteonecrosis in vitro, as well as to elucidate if VEGF mediating vascular permeability and osteoclastic bone resorption are Src dependent. METHODS We employed pharmacological VEGF to induce higher endothelial permeability and osteoclasts activity for simulating related pathological features of destructive repair in vitro. Src specific pp60(c-src)siRNA was used for determining the contribution of VEGF and Src to destructive repair. The primary endothelial cells and osteoclasts were treated with 50ng/ml VEGF and/or transfected with the pp60(c-src)siRNA, while equivalent PBS and non-targeting siRNA were treated in the control groups. RESULTS VEGF enhanced Src bioactivity through promoting dephosphorylation of Src at Y527 and phosphorylation of Src at Y416. Meanwhile, Src specific pp60(c-src)siRNA significantly reduced Src expression in both cells. VEGF destroyed the junctional integrity of endothelial cells resulting in higher endothelial permeability. However, Src blockade significantly relieved VEGF induced actin stress and inhibited caveolae and VVOs formation, meanwhile further stabilized the complex β-catenin/VE-cadherin/Flk-1 through decreasing phosphorylation of VE-cadherin, ultimately decreasing VEGF-mediating higher vascular permeability. In addition, VEGF promoted osteoclasts formation and function without affecting the adhesion activity and cytoskeleton. We further found that Src blockade significantly impaired cytoskeleton resulting in a lower adhesion activity through down-regulation of phosphorylation of Src, Pyk2 and Cbl, and ultimately inhibited osteoclasts formation and function. CONCLUSIONS These findings provide a new insight into VEGF and c-Src mode of reaction in triggering destructive repair of osteonecrosis and further indicate that VEGF mediating vascular permeability and osteoclasts activity are Src-dependent. Blockade of Src may have great potential as an effective therapy targeting destructive repair in osteonecrosis.
Collapse
Affiliation(s)
- Hui-Juan Cao
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| | - Li-Zhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - Nan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| | - Lin-Ying Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China; Nano Science and Technology Institute, The University of Science and Technology of China, Suzhou, PR China.
| | - Ye Li
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| | - Dan Li
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| | - Yu-Xiao Lai
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| | - Xin-Luan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Shenzhen Bioactive Materials Engineering Lab for Medicine, Shenzhen, PR China.
| |
Collapse
|
36
|
Cruet-Hennequart S, Drougard C, Shaw G, Legendre F, Demoor M, Barry F, Lefaix JL, Galéra P. Radiation-induced alterations of osteogenic and chondrogenic differentiation of human mesenchymal stem cells. PLoS One 2015; 10:e0119334. [PMID: 25837977 PMCID: PMC4383487 DOI: 10.1371/journal.pone.0119334] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 01/13/2015] [Indexed: 12/27/2022] Open
Abstract
While human mesenchymal stem cells (hMSCs), either in the bone marrow or in tumour microenvironment could be targeted by radiotherapy, their response is poorly understood. The oxic effects on radiosensitivity, cell cycle progression are largely unknown, and the radiation effects on hMSCs differentiation capacities remained unexplored. Here we analysed hMSCs viability and cell cycle progression in 21% O2 and 3% O2 conditions after medical X-rays irradiation. Differentiation towards osteogenesis and chondrogenesis after irradiation was evaluated through an analysis of differentiation specific genes. Finally, a 3D culture model in hypoxia was used to evaluate chondrogenesis in conditions mimicking the natural hMSCs microenvironment. The hMSCs radiosensitivity was not affected by O2 tension. A decreased number of cells in S phase and an increase in G2/M were observed in both O2 tensions after 16 hours but hMSCs released from the G2/M arrest and proliferated at day 7. Osteogenesis was increased after irradiation with an enhancement of mRNA expression of specific osteogenic genes (alkaline phosphatase, osteopontin). Osteoblastic differentiation was altered since matrix deposition was impaired with a decreased expression of collagen I, probably through an increase of its degradation by MMP-3. After induction in monolayers, chondrogenesis was altered after irradiation with an increase in COL1A1 and a decrease in both SOX9 and ACAN mRNA expression. After induction in a 3D culture in hypoxia, chondrogenesis was altered after irradiation with a decrease in COL2A1, ACAN and SOX9 mRNA amounts associated with a RUNX2 increase. Together with collagens I and II proteins decrease, associated to a MMP-13 expression increase, these data show a radiation-induced impairment of chondrogenesis. Finally, a radiation-induced impairment of both osteogenesis and chondrogenesis was characterised by a matrix composition alteration, through inhibition of synthesis and/or increased degradation. Alteration of osteogenesis and chondrogenesis in hMSCs could potentially explain bone/joints defects observed after radiotherapy.
Collapse
Affiliation(s)
- Séverine Cruet-Hennequart
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), Caen, France
- Laboratoire Accueil en Radiobiologie avec les Ions Accélérés (CEA-DSV-IRCM-LARIA), Bd Becquerel, Caen Cedex 5, France
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Carole Drougard
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), Caen, France
| | - Georgina Shaw
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Florence Legendre
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), Caen, France
| | - Magali Demoor
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), Caen, France
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Jean-Louis Lefaix
- Laboratoire Accueil en Radiobiologie avec les Ions Accélérés (CEA-DSV-IRCM-LARIA), Bd Becquerel, Caen Cedex 5, France
| | - Philippe Galéra
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), Caen, France
- * E-mail:
| |
Collapse
|
37
|
Baker N, Boyette LB, Tuan RS. Characterization of bone marrow-derived mesenchymal stem cells in aging. Bone 2015; 70:37-47. [PMID: 25445445 DOI: 10.1016/j.bone.2014.10.014] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 12/17/2022]
Abstract
Adult mesenchymal stem cells are a resource for autologous and allogeneic cell therapies for immune-modulation and regenerative medicine. However, patients most in need of such therapies are often of advanced age. Therefore, the effects of the aged milieu on these cells and their intrinsic aging in vivo are important considerations. Furthermore, these cells may require expansion in vitro before use as well as for future research. Their aging in vitro is thus also an important consideration. Here, we focus on bone marrow mesenchymal stem cells (BMSCs), which are unique compared to other stem cells due to their support of hematopoietic cells in addition to contributing to bone formation. BMSCs may be sensitive to age-related diseases and could perpetuate degenerative diseases in which bone remodeling is a contributory factor. Here, we review (1) the characterization of BMSCs, (2) the characterization of in vivo-aged BMSCs, (3) the characterization of in vitro-aged BMSCs, and (4) potential approaches to optimize the performance of aged BMSCs. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Natasha Baker
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa B Boyette
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
38
|
Flowers S, Patel PJ, Gleicher S, Amer K, Himelman E, Goel S, Moran E. p107-Dependent recruitment of SWI/SNF to the alkaline phosphatase promoter during osteoblast differentiation. Bone 2014; 69:47-54. [PMID: 25182511 PMCID: PMC5222550 DOI: 10.1016/j.bone.2014.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 08/11/2014] [Accepted: 08/16/2014] [Indexed: 12/28/2022]
Abstract
The retinoblastoma protein family is intimately involved in the regulation of tissue specific gene expression during mesenchymal stem cell differentiation. The role of the following proteins, pRB, p107 and p130, is particularly significant in differentiation to the osteoblast lineage, as human germ-line mutations of RB1 greatly increase susceptibility to osteosarcoma. During differentiation, pRB directly targets certain osteogenic genes for activation, including the alkaline phosphatase-encoding gene Alpl. Chromatin immunoprecipitation (ChIP) assays indicate that Alpl is targeted by p107 in differentiating osteoblasts selectively during activation with the same dynamics as pRB, which suggests that p107 helps promote Alpl activation. Mouse models indicate overlapping roles for pRB and p107 in bone and cartilage formation, but very little is known about direct tissue-specific gene targets of p107, or the consequences of targeting by p107. Here, the roles of p107 and pRB were compared using shRNA-mediated knockdown genetics in an osteoblast progenitor model, MC3T3-E1 cells. The results show that p107 has a distinct role along with pRB in induction of Alpl. Deficiency of p107 does not impede recruitment of transcription factors recognized as pRB co-activation partners at the promoter; however, p107 is required for the efficient recruitment of an activating SWI/SNF chromatin-remodeling complex, an essential event in Alpl induction.
Collapse
Affiliation(s)
- Stephen Flowers
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA
| | - Parth J Patel
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA
| | - Stephanie Gleicher
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA
| | - Kamal Amer
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA
| | - Eric Himelman
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA
| | - Shruti Goel
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA
| | - Elizabeth Moran
- Department of Orthopaedics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ 07103, USA.
| |
Collapse
|
39
|
Immunosuppressive capabilities of mesenchymal stromal cells are maintained under hypoxic growth conditions and after gamma irradiation. Cytotherapy 2014; 17:152-62. [PMID: 25453724 DOI: 10.1016/j.jcyt.2014.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 09/29/2014] [Accepted: 10/07/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS The discovery of regenerative and immunosuppressive capacities of mesenchymal stromal cells (MSCs) raises hope for patients with tissue-damaging or severe, treatment-refractory autoimmune disorders. We previously presented a method to expand human MSCs in a bioreactor under standardized Good Manufacturing Practice conditions. Now we characterized the impact of critical treatment conditions on MSCs with respect to immunosuppressive capabilities and proliferation. METHODS MSC proliferation and survival after γ irradiation were determined by 5-carboxyfluorescein diacetate N-succinimidyl ester and annexinV/4',6-diamidino-2-phenylindole (DAPI) staining, respectively. T-cell proliferation assays were used to assess the effect of γ irradiation, passaging, cryopreservation, post-thaw equilibration time and hypoxia on T-cell suppressive capacities of MSCs. Quantitative polymerase chain reaction and β-galactosidase staining served as tools to investigate differences between immunosuppressive and non-immunosuppressive MSCs. RESULTS γ irradiation of MSCs abrogated their proliferation while vitality and T-cell inhibitory capacity were preserved. Passaging and long cryopreservation time decreased the T-cell suppressive function of MSCs, and postthaw equilibration time of 5 days restored this capability. Hypoxic culture markedly increased MSC proliferation without affecting their T-cell-suppressive capacity and phenotype. Furthermore, T-cell suppressive MSCs showed higher CXCL12 expression and less β-galactosidase staining than non-suppressive MSCs. DISCUSSION We demonstrate that γ irradiation is an effective strategy to abrogate MSC proliferation without impairing the cells' immunosuppressive function. Hypoxia significantly enhanced MSC expansion, allowing for transplantation of MSCs with low passage number. In summary, our optimized MSC expansion protocol successfully addressed the issues of safety and preservation of immunosuppressive MSC function after ex vivo expansion for therapeutic purposes.
Collapse
|
40
|
Lin SP, Chiu FY, Wang Y, Yen ML, Kao SY, Hung SC. RB maintains quiescence and prevents premature senescence through upregulation of DNMT1 in mesenchymal stromal cells. Stem Cell Reports 2014; 3:975-86. [PMID: 25455074 PMCID: PMC4264040 DOI: 10.1016/j.stemcr.2014.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 10/05/2014] [Accepted: 10/06/2014] [Indexed: 12/14/2022] Open
Abstract
Many cell therapies currently being tested are based on mesenchymal stromal cells (MSCs). However, MSCs start to enter the senescent state upon long-term expansion. The role of retinoblastoma (RB) protein in regulating MSC properties is not well studied. Here, we show that RB levels are higher in early-passage MSCs compared with late-passage MSCs. RB knockdown induces premature senescence and reduced differentiation potentials in early-passage MSCs. RB overexpression inhibits senescence and increases differentiation potentials in late-passage MSCs. Expression of DNMT1, but not DNMT3A or DNMT3B, is also higher in early-passage MSCs than in late-passage MSCs. Furthermore, DNMT1 knockdown in early-passage MSCs induces senescence and reduces differentiation potentials, whereas DNMT1 overexpression in late-passage MSCs has the opposite effect. These results demonstrate that RB expressed in early-passage MSCs upregulates DNMT1 expression and inhibits senescence in MSCs. Therefore, genetic modification of RB could be a way to improve the efficiency of MSCs in clinical use.
Collapse
Affiliation(s)
- Shih-Pei Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan, ROC
| | - Fang-Yao Chiu
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 112, Taiwan, ROC
| | - Yu Wang
- Department of Dentistry Sciences, National Yang-Ming University, Taipei 112, Taiwan, ROC; Department of Stomatology, Taipei Veterans General Hospital, Taipei 112, Taiwan, ROC
| | - Men-Luh Yen
- Departments of Primary Care Medicine and Obstetrics/Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Shou-Yen Kao
- Department of Dentistry Sciences, National Yang-Ming University, Taipei 112, Taiwan, ROC; Department of Stomatology, Taipei Veterans General Hospital, Taipei 112, Taiwan, ROC.
| | - Shih-Chieh Hung
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan, ROC; Institute of Pharmacology, National Yang-Ming University, Taipei 112, Taiwan, ROC; Institute of Traditional Medicine, Faculty of Medicine, National Yang-Ming University, Taipei 112, Taiwan, ROC; Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 112, Taiwan, ROC; Stem Cell Laboratory, Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei 112, Taiwan, ROC; Institute of Biomedical Sciences, Academia Sinica, Taipei 105, Taiwan, ROC.
| |
Collapse
|
41
|
Hwang ES. Senescence suppressors: their practical importance in replicative lifespan extension in stem cells. Cell Mol Life Sci 2014; 71:4207-19. [PMID: 25052377 PMCID: PMC11113678 DOI: 10.1007/s00018-014-1685-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/23/2014] [Accepted: 07/14/2014] [Indexed: 02/06/2023]
Abstract
Recent animal and clinical studies report promising results for the therapeutic utilization of stem cells in regenerative medicine. Mesenchymal stem cells (MSCs), with their pluripotent nature, have advantages over embryonic stem cells in terms of their availability and feasibility. However, their proliferative activity is destined to slow by replicative senescence, and the limited proliferative potential of MSCs not only hinders the preparation of sufficient cells for in vivo application, but also draws a limitation on their potential for differentiation. This calls for the development of safe and efficient means to increase the proliferative as well as differentiation potential of MSCs. Recent advances have led to a better understanding of the underlying mechanisms and significance of cellular senescence, facilitating ways to manipulate the replicative lifespan of a variety of primary cells, including MSCs. This paper introduces a class of proteins that function as senescence suppressors. Like tumor suppressors, these proteins are lost in senescence, while their forced expression delays the onset of senescence. Moreover, treatments that increase the expression or the activity of senescence suppressors, therefore, cause expansion of the replicative and differentiation potential of MSCs. The nature of the activities and putative underlying mechanisms of the senescence suppressors will be discussed to facilitate their evaluation.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemungu, Jeonnongdong 90, Seoul, 130-743, Republic of Korea,
| |
Collapse
|
42
|
MiR-101 induces senescence and prevents apoptosis in the background of DNA damage in MCF7 cells. PLoS One 2014; 9:e111177. [PMID: 25353636 PMCID: PMC4213038 DOI: 10.1371/journal.pone.0111177] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022] Open
Abstract
Moderately increased DNA damage due to the exogenous miR-101 (4 fold) over-expression in MCF7 cells was substantiated by an increase in the number of γ-H2AX foci, correlating with a simple-to-do Halo-assay. miR-101 induced mild/moderate DNA damage favoured senescence rather than apoptosis. An experimental support emanated from the induced mild/moderate DNA damage with 1 µM/5 µM etoposide in MCF7 cells, which resulted in an endogenous miR-101 over-expression (10/4 fold, respectively), followed by senescence. On the other hand, the severe DNA damage induced with 10 µM etoposide, resulted in a low (<1 fold) endogenous expression of miR-101 and an elevated percentage of apoptotic cells. Using bioinformatics tools along with in-vitro and in-vivo validations, miR-101 was found to target and downregulate the mRNA expression of UBE2N and SMARCA4, involved in DNA damage repair (DDR) pathways. Recovery of the expression of the two novel targets in anti-miR-101 transfection validated the results. We conclude that a threshold range of over-expressed miR-101, capable of inducing mild/moderate DNA damage, is sensed by cells to become senescent. The observation derives further support from in-silico protein-protein network analysis where the two novel targets showed their involvement in senescence pathway.
Collapse
|
43
|
Galderisi U, Giordano A. The gap between the physiological and therapeutic roles of mesenchymal stem cells. Med Res Rev 2014; 34:1100-26. [PMID: 24866817 DOI: 10.1002/med.21322] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Several investigators have cultivated marrow stromal cells and have identified a population of mesenchymal stem cells (MSCs). These cells expand extensively in vitro and exhibit multilineage differentiation potential. The lack of MSC-specific markers impedes identification of MSC functions. Further in vivo studies of these cells may elucidate the nature of MSCs. Although the nature of MSCs remains unclear, nonclonal stromal cultures are used as a source of putative MSCs for therapeutic purposes. Preclinical studies and clinical trials assumed that transplanted MSCs exert their effects through their differentiation properties or through the release of molecules that restore tissue functions and modulate immune cells. These studies reported contradictory results and failed to meet expectations. Thus, it is important to note that current protocols for MSC therapy are primarily based on the use of in vitro expanded nonclonal MSCs. Clearly defining the physiological features of in situ MSCs and the in vitro and in vivo properties of nonclonal cultures of stromal cells, which are often misidentified as pure stem cell cultures, may explain the reported failures of MSC therapy. This review will address these issues.
Collapse
Affiliation(s)
- Umberto Galderisi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania; Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy; Genkök Stem Cell Centre, Erciyes University, Kayseri, Turkey
| | | |
Collapse
|
44
|
Schellenberg A, Mauen S, Koch CM, Jans R, de Waele P, Wagner W. Proof of principle: quality control of therapeutic cell preparations using senescence-associated DNA-methylation changes. BMC Res Notes 2014; 7:254. [PMID: 24755407 PMCID: PMC4005405 DOI: 10.1186/1756-0500-7-254] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 04/11/2014] [Indexed: 12/31/2022] Open
Abstract
Background Tracking of replicative senescence is of fundamental relevance in cellular therapy. Cell preparations – such as mesenchymal stromal cells (MSCs) - undergo continuous changes during culture expansion, which is reflected by impaired proliferation and loss of differentiation potential. This process is associated with epigenetic modifications: during in vitro culture, cells acquire senescence-associated DNA methylation (SA-DNAm) changes at specific sites in the genome. We have recently described an Epigenetic-Senescence-Signature that facilitates prediction of the state of cellular aging by analysis of DNAm at six CpG sites (associated with the genes GRM7, CASR, PRAMEF2, SELP, CASP14 and KRTAP13-3), but this has not yet been proven over subsequent passages and with MSCs isolated under good manufacturing practice (GMP) conditions. Findings MSCs were isolated from human bone marrow and GMP-conform expanded for up to 11 passages. Cumulative population doublings (cPDs) and long-term growth curves were calculated based on cell numbers at each passage. Furthermore, 32 cryopreserved aliquots of these cell preparations were retrospectively analyzed using our Epigenetic-Senescence-Signature: DNAm-level was analyzed at six specific CpGs, and the results were used to estimate cPDs, time of culture expansion, and passage numbers. Overall, predicted and real parameters revealed a good correlation, particularly in cPDs. Based on predicted cPDs we could reconstruct long-term growth curves and demonstrated the continuous increase in replicative senescence on molecular level. Conclusion Epigenetic analysis of specific CpG sites in the genome can be used to estimate the state of cellular aging for quality control of therapeutic cell products.
Collapse
Affiliation(s)
| | | | | | | | | | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Technology, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074 Aachen, Germany.
| |
Collapse
|
45
|
Honoki K, Tsujiuchi T. Senescence bypass in mesenchymal stem cells: a potential pathogenesis and implications of pro-senescence therapy in sarcomas. Expert Rev Anticancer Ther 2014; 13:983-96. [PMID: 23984899 DOI: 10.1586/14737140.2013.820010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cellular senescence is a mechanism that limits the lifespan of somatic cells as the results of replicative proliferation and response to stresses, and that prevents undesired oncogenic changes constituting a barrier against immortalization and tumorigenesis. Mesenchymal stem cells (MSCs) reside in a variety of tissues, and participates in tissue maintenance with their multipotent differentiation ability. MSCs are also considered to be as cells of origin for certain type of sarcomas. We reviewed the mechanisms of cellular senescence in MSCs and hypothesized senescence bypass as the potential pathogenesis for sarcoma development, and proposed the possibility of senescence induction therapy for an alternative treatment strategy against sarcomas, especially cells with the resistance to conventional chemo and radiotherapy including sarcoma stem cells.
Collapse
Affiliation(s)
- Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | |
Collapse
|
46
|
p19INK4d is involved in the cellular senescence mechanism contributing to heterochromatin formation. Biochim Biophys Acta Gen Subj 2014; 1840:2171-83. [PMID: 24667034 DOI: 10.1016/j.bbagen.2014.03.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/26/2014] [Accepted: 03/11/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND During evolution, organisms with renewable tissues have developed mechanisms to prevent tumorigenesis, including cellular senescence and apoptosis. Cellular senescence is characterized by a permanent cell cycle arrest triggered by both endogenous stress and exogenous stress. The p19INK4d, a member of the family of cyclin-dependent kinase inhibitors (INK4), plays an important role on cell cycle regulation and in the cellular DNA damage response. We hypothesize that p19INK4d is a potential factor involved in the onset and/or maintenance of the senescent state. METHODS Senescence was confirmed by measuring the cell cycle arrest and the senescence-associated β-galactosidase activity. Changes in p19INK4d expression and localization during senescence were determined by Western blot and immunofluorescence assays. Chromatin condensation was measured by microccocal nuclease digestion and histone salt extraction. RESULTS The data presented here show for the first time that p19INK4d expression is up-regulated by different types of senescence. Changes in senescence-associated hallmarks were driven by modulation of p19 expression indicating a direct link between p19INK4d induction and the establishment of cellular senescence. Following a senescence stimulus, p19INK4d translocates to the nucleus and tightly associates with chromatin. Moreover, reduced levels of p19INK4d impair senescence-related global genomic heterochromatinization. Analysis of p19INK4d mRNA and protein levels in tissues from differently aged mice revealed an up-regulation of p19INK4d that correlates with age. CONCLUSION We propose that p19INK4d participates in the cellular mechanisms that trigger senescence by contributing to chromatin compaction. GENERAL SIGNIFICANCE This study provides novel insights into the dynamics process of cellular senescence, a central tumor suppressive mechanism.
Collapse
|
47
|
|
48
|
Nicolay NH, Sommer E, Lopez R, Wirkner U, Trinh T, Sisombath S, Debus J, Ho AD, Saffrich R, Huber PE. Mesenchymal Stem Cells Retain Their Defining Stem Cell Characteristics After Exposure to Ionizing Radiation. Int J Radiat Oncol Biol Phys 2013; 87:1171-8. [DOI: 10.1016/j.ijrobp.2013.09.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/16/2013] [Accepted: 09/03/2013] [Indexed: 10/26/2022]
|
49
|
Capasso S, Alessio N, Di Bernardo G, Cipollaro M, Melone MA, Peluso G, Giordano A, Galderisi U. Silencing of RB1 and RB2/P130 during adipogenesis of bone marrow stromal cells results in dysregulated differentiation. Cell Cycle 2013; 13:482-90. [PMID: 24281253 DOI: 10.4161/cc.27275] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Bone marrow adipose tissue (BMAT) is different from fat found elsewhere in the body, and only recently have some of its functions been investigated. BMAT may regulate bone marrow stem cell niche and plays a role in energy storage and thermogenesis. BMAT may be involved also in obesity and osteoporosis onset. Given the paramount functions of BMAT, we decided to better clarify the human bone marrow adipogenesis by analyzing the role of the retinoblastoma gene family, which are key players in cell cycle regulation. Our data provide evidence that the inactivation of RB1 or RB2/P130 in uncommitted bone marrow stromal cells (BMSC) facilitates the first steps of adipogenesis. In cultures with silenced RB1 or RB2/P130, we observed an increase of clones with adipogenic potential and a higher percentage of cells accumulating lipid droplets. Nevertheless, the absence of RB1 or RB2/P130 impaired the terminal adipocyte differentiation and gave rise to dysregulated adipose cells, with alteration in lipid uptake and release. For the first time, we evidenced that RB2/P130 plays a role in bone marrow adipogenesis. Our data suggest that while the inactivation of retinoblastoma proteins may delay the onset of last cell division and allow more BMSC to be committed to adipocyte, it did not allow a permanent cell cycle exit, which is a prerequisite for adipocyte terminal maturation.
Collapse
Affiliation(s)
- Stefania Capasso
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy; Institute of Protein Biochemistry; CNR; Naples, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy
| | - Mariarosa Ab Melone
- Department of Clinical and Experimental Medicine; Second University of Naples; Naples, Italy
| | - Gianfranco Peluso
- Institute of Protein Biochemistry; CNR; Naples, Italy; Institute of Biomedicine and Bioresources; CNR; Naples, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine; Center For Biotechnology; Temple University; Philadelphia, PA USA; Department of Medicine, Surgery and Neurociences; University of Siena; Siena, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine; Biotechnology and Molecular Biology Section; Second University of Naples; Naples, Italy; Institute of Protein Biochemistry; CNR; Naples, Italy; Sbarro Institute for Cancer Research and Molecular Medicine; Center For Biotechnology; Temple University; Philadelphia, PA USA; GENKÖK; Genome and Stem Cell Center; Erciyes University; Kayseri, Turkey
| |
Collapse
|
50
|
Severino V, Alessio N, Farina A, Sandomenico A, Cipollaro M, Peluso G, Galderisi U, Chambery A. Insulin-like growth factor binding proteins 4 and 7 released by senescent cells promote premature senescence in mesenchymal stem cells. Cell Death Dis 2013; 4:e911. [PMID: 24201810 PMCID: PMC3847322 DOI: 10.1038/cddis.2013.445] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/31/2013] [Accepted: 09/12/2013] [Indexed: 01/05/2023]
Abstract
Cellular senescence is the permanent arrest of cell cycle, physiologically related to aging and aging-associated diseases. Senescence is also recognized as a mechanism for limiting the regenerative potential of stem cells and to protect cells from cancer development. The senescence program is realized through autocrine/paracrine pathways based on the activation of a peculiar senescence-associated secretory phenotype (SASP). We show here that conditioned media (CM) of senescent mesenchymal stem cells (MSCs) contain a set of secreted factors that are able to induce a full senescence response in young cells. To delineate a hallmark of stem cells SASP, we have characterized the factors secreted by senescent MSC identifying insulin-like growth factor binding proteins 4 and 7 (IGFBP4 and IGFBP7) as key components needed for triggering senescence in young MSC. The pro-senescent effects of IGFBP4 and IGFBP7 are reversed by single or simultaneous immunodepletion of either proteins from senescent-CM. The blocking of IGFBP4/7 also reduces apoptosis and promotes cell growth, suggesting that they may have a pleiotropic effect on MSC biology. Furthermore, the simultaneous addition of rIGFBP4/7 increased senescence and induced apoptosis in young MSC. Collectively, these results suggest the occurrence of novel-secreted factors regulating MSC cellular senescence of potential importance for regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- V Severino
- 1] Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Caserta, Italy [2] Institute of Biostructures and Bioimaging-IBB, CNR, Napoli, Italy [3] Centro Interuniversitario di Ricerca sui Peptidi Bioattivi-CIRPEB, Napoli, Italy
| | | | | | | | | | | | | | | |
Collapse
|