1
|
Mazzella M, Walker K, Cormier C, Kapanowski M, Ishmakej A, Saifee A, Govind Y, Chaudhry GR. Regulation of self-renewal and senescence in primitive mesenchymal stem cells by Wnt and TGFβ signaling. Stem Cell Res Ther 2023; 14:305. [PMID: 37880755 PMCID: PMC10601332 DOI: 10.1186/s13287-023-03533-y] [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: 01/24/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND The therapeutic application of multipotent mesenchymal stem cells (MSCs) encounters significant challenges, primarily stemming from their inadequate growth and limited self-renewal capabilities. Additionally, as MSCs are propagated, their ability to self-renew declines, and the exact cellular and molecular changes responsible for this are poorly understood. This study aims to uncover the complex molecular mechanisms that govern the self-renewal of primitive (p) MSCs. METHODS We grew pMSCs using two types of medium, fetal bovine serum (FM) and xeno-free (XM), at both low passage (LP, P3) and high passage (HP, P20). To evaluate LP and HP pMSCs, we examined their physical characteristics, cell surface markers, growth rate, colony-forming ability, BrdU assays for proliferation, telomerase activity, and potential to differentiate into three lineages. Moreover, we conducted RNA-seq to analyze their transcriptome and MNase-seq analysis to investigate nucleosome occupancies. RESULTS When grown in FM, pMSCs underwent changes in their cellular morphology, becoming larger and elongated. This was accompanied by a decrease in the expression of CD90 and CD49f, as well as a reduction in CFE, proliferation rate, and telomerase activity. In addition, these cells showed an increased tendency to differentiate into the adipogenic lineage. However, when grown in XM, pMSCs maintained their self-renewal capacity and ability to differentiate into multiple lineages while preserving their fibroblastoid morphology. Transcriptomic analysis showed an upregulation of genes associated with self-renewal, cell cycle regulation, and DNA replication in XM-cultured pMSCs, while senescence-related genes were upregulated in FM-cultured cells. Further analysis demonstrated differential nucleosomal occupancies in self-renewal and senescence-related genes for pMSCs grown in XM and FM, respectively. These findings were confirmed by qRT-PCR analysis, which revealed alterations in the expression of genes related to self-renewal, cell cycle regulation, DNA replication, differentiation, and senescence. To understand the underlying mechanisms, we investigated the involvement of Wnt and TGFβ signaling pathways by modulating them with agonists and antagonists. This experimental manipulation led to the upregulation and downregulation of self-renewal genes in pMSCs, providing further insights into the signaling pathways governing the self-renewal and senescence of pMSCs. CONCLUSION Our study shows that the self-renewal potential of pMSCs is associated with the Wnt pathway, while senescence is linked to TGFβ.
Collapse
Affiliation(s)
- Matteo Mazzella
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Keegan Walker
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Christina Cormier
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Michael Kapanowski
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Albi Ishmakej
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Azeem Saifee
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Yashvardhan Govind
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA.
| |
Collapse
|
2
|
Mazzella M, Walker K, Cormier C, Kapanowski M, Ishmakej A, Saifee A, Govind Y, Chaudhry GR. WNT and VEGF/PDGF signaling regulate self-renewal in primitive mesenchymal stem cells. RESEARCH SQUARE 2023:rs.3.rs-2512048. [PMID: 37090660 PMCID: PMC10120760 DOI: 10.21203/rs.3.rs-2512048/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Background Therapeutic use of multipotent mesenchymal stem cells (MSCs) is hampered due to poor growth and limited self-renewal potential. The self-renewal potential of MSCs is also affected during propagation and changes are poorly understood. This study investigated the molecular mechanism involved in the self-renewal of primitive (p) MSCs. Methods pMSCs were cultured to low passage (LP), P3, and high passage (HP), P20, in fetal bovine serum medium (FM) and xeno-free medium (XM). The characteristics of LP and HP pMSCs were evaluated for morphology, expression of cell surface markers, doubling time (DT), colony forming efficiency (CFE), proliferation by BrdU assay, telomerase activity and trilineage differentiation. We then examined transcriptome and nucleosome occupancies using RNA-seq and MNase-seq, respectively analyses. Results pMSCs grown in FM gradually changed morphology to large elongated cells and showed a significant reduction in the expression of CD90 and CD49f, CFE, proliferation, and telomerase activity. In addition, cells had a greater propensity to differentiate into the adipogenic lineage. In contrast, pMSCs grown in XM maintained small fibroblastoid morphology, self-renewal, and differentiation potential. Transcriptomic analysis showed upregulation of genes involved in self-renewal, cell cycle, and DNA replication in XM-grown pMSCs. Whereas senescence genes were upregulated in cells in FM. MNase-seq analysis revealed less nucleosomal occupancies in self-renewal genes and senescence genes in pMSCs grown in XM and FM, respectively. The expression of selected genes associated with self-renewal, cell cycle, DNA replication, differentiation, and senescence was confirmed by qRT-PCR. These results led us to propose signaling pathways involved in the self-renewal and senescence of pMSCs. Conclusion We conclude that the self-renewal potential of pMSCs is controlled by WNT and VEGF/PDGF, but TGFβ and PI3K signaling induce senescence.
Collapse
|
3
|
Lupatov AY, Yarygin KN. Telomeres and Telomerase in the Control of Stem Cells. Biomedicines 2022; 10:biomedicines10102335. [PMID: 36289597 PMCID: PMC9598777 DOI: 10.3390/biomedicines10102335] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Stem cells serve as a source of cellular material in embryogenesis and postnatal growth and regeneration. This requires significant proliferative potential ensured by sufficient telomere length. Telomere attrition in the stem cells and their niche cells can result in the exhaustion of the regenerative potential of high-turnover organs, causing or contributing to the onset of age-related diseases. In this review, stem cells are examined in the context of the current telomere-centric theory of cell aging, which assumes that telomere shortening depends not just on the number of cell doublings (mitotic clock) but also on the influence of various internal and external factors. The influence of the telomerase and telomere length on the functional activity of different stem cell types, as well as on their aging and prospects of use in cell therapy applications, is discussed.
Collapse
|
4
|
He J, You D, Li Q, Wang J, Ding S, He X, Zheng H, Ji Z, Wang X, Ye X, Liu C, Kang H, Xu X, Xu X, Wang H, Yu M. Osteogenesis-Inducing Chemical Cues Enhance the Mechanosensitivity of Human Mesenchymal Stem Cells for Osteogenic Differentiation on a Microtopographically Patterned Surface. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200053. [PMID: 35373921 PMCID: PMC9165486 DOI: 10.1002/advs.202200053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/25/2022] [Indexed: 05/13/2023]
Abstract
Mechanical cues are widely used for regulating cell behavior because of their overarching, extensive, and non-invasive advantages. However, unlike chemical cues, mechanical cues are not efficient enough to determine cell fate independently and improving the mechanosensitivity of cells is rather challenging. In this study, the combined effect of chemical and mechanical cues on the osteogenic differentiation of human mesenchymal stem cells is examined. These results show that chemical cues such as the presence of an osteogenic medium, induce cells to secrete more collagen, and induce integrin for recruiting focal adhesion proteins that mature and cascade a series of events with the help of the mechanical force of the scaffold material. High-resolution, highly ordered hollow-micro-frustum-arrays using double-layer lithography, combined with modified methacrylate gelatin loaded with pre-defined soluble chemicals to provide both chemical and mechanical cues to cells. This approach ultimately facilitates the achievement of cellular osteodifferentiation and enhances bone repair efficiency in a model of femoral fracture in vivo in mice. Moreover, the results also reveal these pivotal roles of Integrin α2/Focal adhesion kinase/Ras homolog gene family member A/Large Tumor Suppressor 1/Yes-associated protein in human mesenchymal stem cells osteogenic differentiation both in vitro and in vivo. Overall, these results show that chemical cues enhance the microtopographical sensitivity of cells.
Collapse
Affiliation(s)
- Jianxiang He
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Dongqi You
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Qi Li
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Jiabao Wang
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Sijia Ding
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Xiaotong He
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Haiyan Zheng
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Zhenkai Ji
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Xia Wang
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Xin Ye
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Chao Liu
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Hanyue Kang
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Xiuzhen Xu
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Xiaobin Xu
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Huiming Wang
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
- School of StomatologyThe First Affiliated Hospital of Zhejiang University School of MedicineHangzhou310003P. R. China
| | - Mengfei Yu
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| |
Collapse
|
5
|
Al Naem M, Bourebaba L, Kucharczyk K, Röcken M, Marycz K. Therapeutic mesenchymal stromal stem cells: Isolation, characterization and role in equine regenerative medicine and metabolic disorders. Stem Cell Rev Rep 2021; 16:301-322. [PMID: 31797146 DOI: 10.1007/s12015-019-09932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a popular treatment modality in equine orthopaedics. Regenerative therapies are especially interesting for pathologies like complicated tendinopathies of the distal limb, osteoarthritis, osteochondritis dissecans (OCD) and more recently metabolic disorders. Main sources for MSC harvesting in the horse are bone marrow, adipose tissue and umbilical cord blood. While the acquisition of umbilical cord blood is fairly easy and non-invasive, extraction of bone marrow and adipose tissue requires more invasive techniques. Characterization of the stem cells as a result of any isolation method, is also a crucial step for the confirmation of the cells' stemness properties; thus, three main characteristics must be fulfilled by these cells, namely: adherence, expression of a series of well-defined differentiation clusters as well as pluripotency. EVs, resulting from the paracrine action of MSCs, also play a key role in the therapeutic mechanisms mediated by stem cells; MSC-EVs are thus largely implicated in the regulation of proliferation, maturation, polarization and migration of various target cells. Evidence that EVs alone represent a complex network 0involving different soluble factors and could then reflect biophysical characteristics of parent cells has fuelled the importance of developing highly specific techniques for their isolation and analysis. All these aspects related to the functional and technical understanding of MSCs will be discussed and summarized in this review.
Collapse
Affiliation(s)
- Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kucharczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
6
|
Lewandowski RB, Stępińska M, Gietka A, Dobrzyńska M, Łapiński MP, Trafny EA. The red-light emitting diode irradiation increases proliferation of human bone marrow mesenchymal stem cells preserving their immunophenotype. Int J Radiat Biol 2021; 97:553-563. [PMID: 33471577 DOI: 10.1080/09553002.2021.1876947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 11/20/2020] [Accepted: 01/05/2021] [Indexed: 01/02/2023]
Abstract
PURPOSE For effective clinical application of human bone marrow mesenchymal stem cells (hBM-MSCs), the enhancement of their proliferation in vitro together with maintaining the expression of their crucial surface antigens and differentiation potential is necessary. The present study aimed to investigate the effect of light-emitting diode (LED) irradiation on hBM-MSCs proliferation after two, five, or nine days post-irradiation. MATERIALS AND METHODS The hBM-MSCs were exposed to the LED light at 630 nm, 4 J/cm2, and power densities of 7, 17, or 30 mW/cm2. To assess the cell proliferation rate in the sham-irradiated and irradiated samples the cells metabolic activity and DNA content were determined. The number of apoptotic and necrotic cells in the samples was also evaluated. The expression of the crucial surface antigens of the hBM-MSCs up to nine days after irradiation at 4 J/cm2 and 17 mW/cm2 was monitored with flow cytometry. Additionally, the potential of hBM-MSCs for induced differentiation was measured. RESULTS When the metabolic activity was assayed, the significant increase in the cell proliferation rate by 31 and 50% after the irradiation with 4 J/cm2 and 17 mW/cm2, respectively, was observed at day five and nine when compared to the sham-irradiated cells (p < .05). Similarly, DNA content within the irradiated hBM-MSCs increased by 31 and 41% at day five and nine after the irradiation with 4 J/cm2 and 17 mW/cm2 in comparison to the sham-irradiated cells. LED irradiation did not change the expression of the crucial surface antigens of the hBM-MSCs up to nine days after irradiation at 4 J/cm2 and 17 mW/cm2. At the same experimental conditions, the hBM-MSCs maintain in vitro their capability for multipotential differentiation into osteoblasts, adipocytes, and chondrocytes. CONCLUSION Therefore, LED irradiation at a wavelength of 630 nm, energy density 4 J/cm2, and power density 17 mW/cm2 can effectively increase the number of viable hBM-MSCs in vitro.
Collapse
Affiliation(s)
- Rafał B Lewandowski
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Małgorzata Stępińska
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Andrzej Gietka
- Optoelectronic Technologies Division, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Monika Dobrzyńska
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Mariusz P Łapiński
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Elżbieta A Trafny
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| |
Collapse
|
7
|
Song SB, Park JS, Chung GJ, Lee IH, Hwang ES. Diverse therapeutic efficacies and more diverse mechanisms of nicotinamide. Metabolomics 2019; 15:137. [PMID: 31587111 DOI: 10.1007/s11306-019-1604-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/30/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Nicotinamide (NAM) is a form of vitamin B3 that, when administered at near-gram doses, has been shown or suggested to be therapeutically effective against many diseases and conditions. The target conditions are incredibly diverse ranging from skin disorders such as bullous pemphigoid to schizophrenia and depression and even AIDS. Similar diversity is expected for the underlying mechanisms. In a large portion of the conditions, NAM conversion to nicotinamide adenine dinucleotide (NAD+) may be a major factor in its efficacy. The augmentation of cellular NAD+ level not only modulates mitochondrial production of ATP and superoxide, but also activates many enzymes. Activated sirtuin proteins, a family of NAD+-dependent deacetylases, play important roles in many of NAM's effects such as an increase in mitochondrial quality and cell viability countering neuronal damages and metabolic diseases. Meanwhile, certain observed effects are mediated by NAM itself. However, our understanding on the mechanisms of NAM's effects is limited to those involving certain key proteins and may even be inaccurate in some proposed cases. AIM OF REVIEW This review details the conditions that NAM has been shown to or is expected to effectively treat in humans and animals and evaluates the proposed underlying molecular mechanisms, with the intention of promoting wider, safe therapeutic application of NAM. KEY SCIENTIFIC CONCEPTS OF REVIEW NAM, by itself or through altering metabolic balance of NAD+ and tryptophan, modulates mitochondrial function and activities of many molecules and thereby positively affects cell viability and metabolic functions. And, NAM administration appears to be quite safe with limited possibility of side effects which are related to NAM's metabolites.
Collapse
Affiliation(s)
- Seon Beom Song
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdae-ro 163, Seoul, Republic of Korea
| | - Jin Sung Park
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdae-ro 163, Seoul, Republic of Korea
| | - Gu June Chung
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdae-ro 163, Seoul, Republic of Korea
| | - In Hye Lee
- Department of Life Science, Ewha Womans University, Ewhayeodae-gil 52, Seoul, Republic of Korea
| | - Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdae-ro 163, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Ok JS, Song SB, Hwang ES. Enhancement of Replication and Differentiation Potential of Human Bone Marrow Stem Cells by Nicotinamide Treatment. Int J Stem Cells 2018; 11:13-25. [PMID: 29699388 PMCID: PMC5984055 DOI: 10.15283/ijsc18033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/31/2022] Open
Abstract
Background and Objectives Therapies using mesenchymal stem cells (MSCs) generally require substantial expansion of cell populations. However, the replicative life span of MSCs is limited and their multipotency declines over continued passages, imposing a limitation on their application especially in aged individuals. In an effort to increase MSC life span, we tested the effects of nicotinamide (NAM), a precursor of NAD+ that has been shown to reduce reactive oxygen species generation and delay the onset of replicative senescence in fibroblasts. Methods Bone marrow stem cells (BMSCs) from healthy donors were cultivated in the presence of 5 mM NAM until the end of their life span. The levels of proliferation and differentiation to osteogenic, adipogenic, and chondrogenic lineages of BMSCs were compared between populations incubated in the absence or presence of NAM. Results The replicative life span was substantially increased with a significant delay in the onset of senescence, and differentiation to all tested lineages was increased. Furthermore, differentiation was sustained and the adipogenic switch from osteogenesis to adipogenesis was attenuated in late-passage BMSCs. Conclusions NAM could be considered as an important biological agent to expand and sustain the multipotency of BMSCs and thus broaden the application of stem cells in cell therapies.
Collapse
Affiliation(s)
- Jeong Su Ok
- Department of Life Science, University of Seoul, Seoul, Korea
| | - Seon Beom Song
- Department of Life Science, University of Seoul, Seoul, Korea
| | - Eun Seong Hwang
- Department of Life Science, University of Seoul, Seoul, Korea
| |
Collapse
|
9
|
Prolonged Growth Hormone/Insulin/Insulin-like Growth Factor Nutrient Response Signaling Pathway as a Silent Killer of Stem Cells and a Culprit in Aging. Stem Cell Rev Rep 2018; 13:443-453. [PMID: 28229284 PMCID: PMC5493720 DOI: 10.1007/s12015-017-9728-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The dream of slowing down the aging process has always inspired mankind. Since stem cells are responsible for tissue and organ rejuvenation, it is logical that we should search for encoded mechanisms affecting life span in these cells. However, in adult life the hierarchy within the stem cell compartment is still not very well defined, and evidence has accumulated that adult tissues contain rare stem cells that possess a broad trans-germ layer differentiation potential. These most-primitive stem cells-those endowed with pluripotent or multipotent differentiation ability and that give rise to other cells more restricted in differentiation, known as tissue-committed stem cells (TCSCs) - are of particular interest. In this review we present the concept supported by accumulating evidence that a population of so-called very small embryonic-like stem cells (VSELs) residing in adult tissues positively impacts the overall survival of mammals, including humans. These unique cells are prevented in vertebrates from premature depletion by decreased sensitivity to growth hormone (GH), insulin (INS), and insulin-like growth factor (IGF) signaling, due to epigenetic changes in paternally imprinted genes that regulate their resistance to these factors. In this context, we can envision nutrient response GH/INS/IGF signaling pathway as a lethal factor for these most primitive stem cells and an important culprit in aging.
Collapse
|
10
|
Barreto S, Gonzalez-Vazquez A, Cameron AR, Cavanagh B, Murray DJ, O'Brien FJ. Identification of the mechanisms by which age alters the mechanosensitivity of mesenchymal stromal cells on substrates of differing stiffness: Implications for osteogenesis and angiogenesis. Acta Biomater 2017; 53:59-69. [PMID: 28216301 DOI: 10.1016/j.actbio.2017.02.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/08/2017] [Accepted: 02/15/2017] [Indexed: 12/21/2022]
Abstract
In order to identify the mechanisms by which skeletal maturity alters the mechanosensitivity of mesenchymal stromal cells (MSCs) and, the implications for osteogenesis and angiogenesis during bone formation, we compared the response of MSCs derived from children and skeletally-mature healthy adults cultured on soft and stiff collagen-coated polyacrylamide substrates. MSCs from children were more mechanosensitive, showing enhanced angiogenesis and osteogenesis on stiff substrates as indicated by increased endothelial tubule formation, PGF production, nuclear-translocation of YAP, ALP activity and mineralisation. To examine these mechanisms in more detail, a customised PCR array identified an age-dependent, stiffness-induced upregulation of NOX1, VEGFR1, VEGFR2, WIF1 and, of particular interest, JNK3 in cells from children compared to adults. When JNK3 activity was inhibited, a reduction in stiffness-induced driven osteogenesis was observed - suggesting that JNK3 might serve as a novel target for recapitulating the enhanced regenerative potential of children in adults suffering from bone degeneration. STATEMENT OF SIGNIFICANCE We investigated the age-associated changes in the capacity of MSCs for bone regeneration involving the mechanosensitive signalling pathways, which reduce the ability of adult cells to respond to biophysical cues in comparison to cells from children, who are still undergoing bone development. Our results offer new insights into the mechanobiology of MSCs and sheds new light on age-altered mechanosensitivity and, on why children have such an immense capacity to regenerate their skeletal system. We have identified the mechanisms by which skeletal maturity alters the mechanosensitivity of mesenchymal stromal cells and an age-dependent, stiffness-induced upregulation of a number of prominent genes including, most notably, JNK3 in children cells, thus suggesting its potential to promote enhanced bone repair.
Collapse
Affiliation(s)
- Sara Barreto
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, RCSI & TCD, Dublin 2, Ireland
| | - Arlyng Gonzalez-Vazquez
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, RCSI & TCD, Dublin 2, Ireland
| | - Andrew R Cameron
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, RCSI & TCD, Dublin 2, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Dylan J Murray
- National Paediatric Craniofacial Centre, Temple Street Children's University Hospital, Dublin 1, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, RCSI & TCD, Dublin 2, Ireland.
| |
Collapse
|