Elsevier

Ageing Research Reviews

Volume 62, September 2020, 101125
Ageing Research Reviews

Review
DNA damage repair response in mesenchymal stromal cells: From cellular senescence and aging to apoptosis and differentiation ability

https://doi.org/10.1016/j.arr.2020.101125Get rights and content

Highlights

Abstract

Mesenchymal stromal cells (MSCs) are heterogeneous and contain several populations, including stem cells. MSCs' secretome has the ability to induce proliferation, differentiation, chemo-attraction, anti-apoptosis, and immunomodulation activities in stem cells. Moreover, these cells recognize tissue damage caused by drugs, radiation (e.g., Ultraviolet, infra-red) and oxidative stress, and respond in two ways: either MSCs differentiate into particular cell lineages to preserve tissue homeostasis, or they release a regenerative secretome to activate tissue repairing mechanisms. The maintenance of MSCs in quiescence can increase the incidence and accumulation of various forms of genomic modifications, particularly upon environmental insults. Thus, dysregulated DNA repair pathways can predispose MSCs to senescence or apoptosis, reducing their stemness and self-renewal properties. For instance, DNA damage can impair telomere replication, activating DNA damage checkpoints to maintain MSC function. In this review, we aim to summarize the role of DNA damage and associated repair responses in MSC senescence, differentiation and programmed cell death.

Introduction

The number and function of stem cells (SCs), in addition to their compartmentalization, contribute to the pathophysiological status of tissues (Vitale et al., 2017a). Adult stem cells (ASCs) can promote tissue repair and regeneration upon injury and sustain inter-cellular heterogeneity for physiological homeostasis. Endogenous and exogenous factors can cause damage onto the DNA and these genetic lesions may challenge ASCs survival and function. The significant roles of DNA in living organisms necessitate precise control of DNA damage repair mechanisms through recruitment of repair factors to damage sites and activation of checkpoint regulators to halt cell-cycle progression. These mechanisms, which are known collectively as the DNA damage response (DDR), can execute full repair or promote the elimination of damaged cells to protect host organisms against possible carcinogenesis (Bielak-Zmijewska et al., 2018). Adult stem and progenitor cells in various tissues are equipped with several regulatory mechanisms to guarantee genome integrity and tissue homeostasis. Dysregulation of DNA repair pathways in SCs can reduce tissue regeneration capacity by limiting the self-renewal and differentiation properties of SCs and by inducing senescence or apoptosis in these cells (Weeden and Asselin-Labat, 2018).

The secretome of MSCs contains tissue repairing elements that play an essential role in regulating local and remote progenitor and stem cell (SC) function. In response to tissue damage, MSCs can release factors that activate tissue repair mechanisms or that direct differentiation of SCs into certain cell lineages (Fitzsimmons et al., 2018; Moravej et al., 2019; Xi et al., 2013). In the present review, we summarize the contributions of the DDR in MSC senescence and apoptosis and highlight its pathophysiological relevance.

Section snippets

Mesenchymal stromal cells

Mesenchymal stromal cells (MSCs) are characterized by fibroblast-like morphology, these cells are derived from bone marrow (BM) and different tissues (Horwitz et al., 2005). The International Society for Cell & Gene Therapy (ISCT) defines MSCs as a heterogeneous population of stromal cells that are capable of self-renewal and tri-lineage differentiation (to osteoblast, adipocyte, and chondrocyte). These cells are plastic-adherent under routine culture conditions and express CD73, CD90, and CD105

Effect of DNA damage in MSCs

In contrast to somatic cells, which typically undergo terminal differentiation, SCs can survive and duplicate for an extended period, which can increase the possibility of incidence and accumulation of damages onto the DNA of these cells (Mani et al., 2019). Chemical reactions between DNA and active molecules, including intracellular reactive oxygen species (ROSs), can cause a wide range of DNA damage (Chatterjee and Walker, 2017). The main endogenous DNA-damages result from metabolic processes

Role of DNA damage in stemness and differentiation of MSCs

Irradiation can trigger ROS accumulation in cells and cause DNA damage and eventually lead to the loss of stemness in MSCs. BM-MSCs show a different response upon exposure to low or high dose radiation. Low dose radiation can induce senescence, a defense response against tumorigenesis, and reduce the stemness of MSCs by attenuating autophagy activity (Alessio et al., 2015). Autophagy is an active mechanism to maintain the stemness of MSCs by decreasing ROS accumulation and DNA damage.

DNA damage in senescence and apoptosis of MSCs

DDR proteins are responsible for the detection of DNA damage as well as for executing the appropriate cellular responses toward repair, senescence or apoptosis (Lombard et al., 2005; Roos and Kaina, 2006). Senescence is a unique state of cell-cycle arrest, which can be induced by various cellular stresses, including DNA damage accumulation. This process is one of the critical defense mechanisms to avoid malignancy in mammalian cells. Although senescent cells are non-replicative, they are

DNA-repair mechanisms in MSCs

Various proteins involved in the DDR and DNA repair recognize DNA damage and either restore DNA integrity or induce senescence, differentiation, or apoptosis in MSCs (Krokan and Bjørås, 2013). In all cells, there are different DNA repair pathways that respond based on the DNA damage type. DNA repair activity is under the influence of many modulators, including epigenetics and other factors, which can regulate the gene expression and post-transcriptional modification (ubiquitination,

Conclusion

MSCs can modulate tissue homeostasis. Also, they can control tissue repair and regeneration capacity in aging-associated degenerative diseases. Although MSCs are located in hypoxic niches to keep away from oxidative stressors and maintain their stemness properties, they are still susceptible to intrinsic and extrinsic DNA-damaging agents. The primary responses of MSCs to DNA damage are the production of a considerable amount of anti-oxidants and activation of the DDR to reduce genotoxicity.

Declaration of Competing Interest

The authors have no conflict of interest to declare relevant to the content of this review.

Acknowledgment

There were no funding resources for the current study.

References (113)

  • E.M. Horwitz et al.

    Clarification of the nomenclature for MSC: the international society for cellular therapy position statement

    Cytotherapy

    (2005)
  • E.E. Kennedy et al.

    Initiating base excision repair in chromatin

    DNA Repair (Amst).

    (2018)
  • D. Liu et al.

    DNA mismatch repair and its many roles in eukaryotic cells

    Mutat. Res. Mutat. Res.

    (2017)
  • D.B. Lombard et al.

    DNA repair, genome stability, and aging

    Cell

    (2005)
  • R. Lopez Perez et al.

    Mesenchymal stem cells preserve their stem cell traits after exposure to antimetabolite chemotherapy

    Stem Cell Res.

    (2019)
  • Y. Lu et al.

    miR675 Accelerates malignant transformation of mesenchymal stem cells by blocking DNA mismatch repair

    Mol. Ther. Nucleic Acids

    (2019)
  • N.K. McGhee et al.

    Elevated corticosterone associated with food deprivation upregulates expression in rat skeletal muscle of the mTORC1 repressor, REDD1

    J. Nutr.

    (2009)
  • J. Munro et al.

    Histone deacetylase inhibitors induce a senescence-like state in human cells by a p16-dependent mechanism that is independent of a mitotic clock

    Exp. Cell Res.

    (2004)
  • V. Natarajan

    Regulation of DNA repair by non-coding miRNAs

    Noncoding RNA Res.

    (2016)
  • N.H. Nicolay et al.

    Mesenchymal stem cells exhibit resistance to topoisomerase inhibition

    Cancer Lett.

    (2016)
  • Y. Pommier et al.

    Repair of topoisomerase I‐Mediated DNA damage

    Progress in Nucleic Acid Research and Molecular Biology

    (2006)
  • W.P. Roos et al.

    DNA damage-induced cell death by apoptosis

    Trends Mol. Med.

    (2006)
  • A. Srinivasan et al.

    Mesenchymal stem cell–derived products for tissue repair and regeneration

    A Roadmap to Non-Hematopoietic Stem Cell-Based Therapeutics

    (2019)
  • J. Thacker et al.

    The mammalian XRCC genes: their roles in DNA repair and genetic stability

    DNA Repair (Amst)

    (2003)
  • J. Thacker et al.

    The XRCC genes: expanding roles in DNA double-strand break repair

    DNA Repair (Amst)

    (2004)
  • A. Tubbs et al.

    Endogenous DNA damage as a source of genomic instability in cancer

    Cell

    (2017)
  • S. Viswanathan et al.

    Mesenchymal stem versus stromal cells: international Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell committee position statement on nomenclature

    Cytotherapy

    (2019)
  • N. Alessio et al.

    Low dose radiation induced senescence of human mesenchymal stromal cells and impaired the autophagy process

    Oncotarget

    (2015)
  • N. Alessio et al.

    Mesenchymal stromal cells having inactivated RB1 survive following low irradiation and accumulate damaged DNA: hints for side effects following radiotherapy

    Cell Cycle

    (2017)
  • H. Alves et al.

    A link between the accumulation of DNA damage and loss of multi-potency of human mesenchymal stromal cells

    J. Cell. Mol. Med.

    (2010)
  • B.N. Ames et al.

    Dietary pesticides (99.99% all natural)

    Proc. Natl. Acad. Sci. U. S. A.

    (1990)
  • S. Aqmasheh et al.

    Effects of mesenchymal stem cell derivatives on hematopoiesis and hematopoietic stem cells

    Adv. Pharm. Bull.

    (2017)
  • F.Z. Asumda et al.

    Age-related changes in rat bone-marrow mesenchymal stem cell plasticity

    BMC Cell Biol.

    (2011)
  • F.Z. Asumda et al.

    Age-related changes in rat bone-marrow mesenchymal stem cell plasticity

    BMC Cell Biol.

    (2011)
  • L. Benameur et al.

    Toward an understanding of mechanism of aging-induced oxidative stress in human mesenchymal stem cells

    Biomed. Mater. Eng.

    (2015)
  • J.W. Bennett et al.

    Mycotoxins

    Clin. Microbiol. Rev.

    (2003)
  • E.H. Blackburn

    Telomere states and cell fates

    Nature

    (2000)
  • S. Bork et al.

    DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells

    Aging Cell

    (2010)
  • A.V. Borodkina et al.

    Different protective mechanisms of human embryonic and endometrium-derived mesenchymal stem cells under oxidative stress

    Cell tissue biol.

    (2014)
  • W. Broekman et al.

    TNF-aα and IL-1β-activated human mesenchymal stromal cells increase airway epithelial wound healing in vitro via activation of the epidermal growth factor receptor

    Respir. Res.

    (2016)
  • A. Caplan

    Why are MSCs therapeutic? New data: new insight

    J. Pathol.

    (2009)
  • N. Chatterjee et al.

    Mechanisms of DNA damage, repair, and mutagenesis

    Environ. Mol. Mutagen.

    (2017)
  • M.A. Chaudhry et al.

    Radiation-induced Micro-RNA modulation in glioblastoma cells differing in DNA-Repair pathways

    DNA Cell Biol.

    (2010)
  • A. Conforti et al.

    Resistance to neoplastic transformation of &i&ex-vivo&/i& expanded human mesenchymal stromal cells after exposure to supramaximal physical and chemical stress

    Oncotarget

    (2016)
  • S. Cruet-Hennequart et al.

    Doxorubicin induces the DNA damage response in cultured human mesenchymal stem cells

    Int. J. Hematol.

    (2012)
  • A. Di Francesco et al.

    The DNA-Damage Response to γ-Radiation Is Affected by miR-27a in A549 Cells

    Int. J. Mol. Sci.

    (2013)
  • A. Di Francesco et al.

    The DNA-Damage Response to γ-radiation is affected by miR-27a in A549 cells

    Int. J. Mol. Sci.

    (2013)
  • K.E.M. Diderich et al.

    Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice

    Age (Omaha)

    (2012)
  • R. Domenis et al.

    Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes

    Sci. Rep.

    (2018)
  • M. Ejtehadifar et al.

    The effects of hypoxia on U937 cell line in mesenchymal stem cells Co-culture system

    Adv. Pharm. Bull.

    (2016)
  • Cited by (36)

    • m6A contributes to a pro-survival state in GC-2 cells by facilitating DNA damage repair: Novel perspectives on the mechanism underlying DEHP genotoxicity in male germ cells

      2023, Science of the Total Environment
      Citation Excerpt :

      The protein level of γH2AX was increased significantly in GC-2 cells exposed to DEHP (Fig. 2 BD), indicating that disrupted genomic integrity during meiosis may be a cause of DEHP-induced spermatogenesis dysfunction. Recent studies have revealed that DNA lesions can cause the collapse of replication forks, a marked reduction in DNA replication activity, and cell cycle arrest (Banimohamad-shotorbani et al., 2020; Roos and Kaina, 2013). To further explore the consequences of DEHP-induced DSBs in GC-2 cells, an EdU assay was performed and DNA replication was examined.

    • Endothelial cells regulated by RNF20 orchestrate the proliferation and differentiation of neural precursor cells during embryonic development

      2022, Cell Reports
      Citation Excerpt :

      However, although the vascular system has important roles in brain development, its functions in NPCs during early brain development remain largely uncharacterized. Previous studies have found that DNA damage in mammalian cells induces a DNA damage response (DDR) that ultimately leads to cell death or cell senescence, depending on the cell type and the extent of DNA damage (Banimohamad-Shotorbani et al., 2020). DNA damage accelerates the telomere shortening and triggers the premature aging of blood vessels.

    • The role of senescence in cellular plasticity: Lessons from regeneration and development and implications for age-related diseases

      2022, Developmental Cell
      Citation Excerpt :

      For example, DNA damage, a known inducer of cellular senescence, triggers the differentiation of some cell types, including keratinocytes (Freije et al., 2014), B cells (Bredemeyer et al., 2008), melanocyte stem cells (Inomata et al., 2009), and others (Sherman et al., 2011; Wingert and Rieger, 2016). Noteworthy, cells showing elevated DNA damage levels exhibit a differentiation bias; for example, damaged hematopoietic stem cells (HSCs) favor differentiation to the myeloid lineage over the lymphoid lineage, whereas damaged mesenchymal stems cells (MSCs) show features of adipocyte differentiation and are less likely to differentiate into osteocytes or chondrocytes (Banimohamad-Shotorbani et al., 2020; Sjakste and Riekstina, 2021). Similar to DNA damage, telomere shortening is also associated with cell differentiation (Forsyth et al., 2002), and telomere dysfunction induces differentiation (Razdan et al., 2018).

    • Senescence in aging

      2022, Molecular, Cellular, and Metabolic Fundamentals of Human Aging
    View all citing articles on Scopus
    1

    These authors are contributed equally to this study.

    View full text