Skip to main content
Journal of Dental Research logoLink to Journal of Dental Research
. 2012 Nov;91(11):1003–1010. doi: 10.1177/0022034512460404

Interplay between Mesenchymal Stem Cells and Lymphocytes

Implications for Immunotherapy and Tissue Regeneration

L Wang 1,2, Y Zhao 1,3, S Shi 1,*
PMCID: PMC3490280  PMID: 22988011

Abstract

In addition to their potential for replacing damaged and diseased tissues by differentiating into tissue-specific cells, mesenchymal stem cells (MSCs) have been found to interact closely with immune cells, such as lymphocytes. In this review, we will discuss current research regarding the immunomodulatory properties of MSCs and the effects of lymphocytes on MSCs. We will suggest how these findings could be translated to potential clinical treatment. MSCs can regulate immune response by inducing activated T-cell apoptosis through the FAS ligand (FASL)/FAS-mediated death pathway via cell-cell contact, leading to up-regulation of regulatory T-cells (Tregs), which ultimately results in immune tolerance. Conversely, lymphocytes can impair survival and osteogenic differentiation of implanted MSCs by secreting the pro-inflammatory cytokines IFN-γ and TNF-α and/or through the FASL/FAS-mediated death pathway, thereby negatively affecting MSC-mediated tissue regeneration. One novel strategy to improve MSC-based tissue engineering involves the reduction of IFN-γ and TNF-α concentration by systemic infusion of Tregs or local application of aspirin. Further understanding of the mechanisms underlying the interplay between lymphocytes and MSCs may be helpful in the development of promising approaches to improve cell-based regenerative medicine and immune therapies.

Keywords: regulatory T-Lymphocytes, immunomodulation, cell therapy, regenerative medicine, interferon-gamma, tumor necrosis factor-alpha

Introduction

Mesenchymal stem cells (MSCs) are a population of plastic-adherent stromal cells with self-renewal and multipotent differentiation capabilities (Friedenstein et al., 1970; Caplan, 1991; Shi and Gronthos, 2003). MSCs reside in a wide spectrum of post-natal tissue types (da Silva Meirelles et al., 2006; Bi et al., 2007), and they have been successfully isolated from several orofacial tissues (Gronthos et al., 2000; Miura et al., 2003; Seo et al., 2004; Morsczeck et al., 2005; Sonoyama et al., 2006; Zhang et al., 2009; Yamaza et al., 2011). In addition to their potential for replacing damaged and diseased tissue by differentiating into tissue-specific cells, MSCs have been shown to interact with hematopoietic stem cells (HSCs) by controlling or directly providing a stem cell niche for HSCs (Sacchetti et al., 2007; Mendez-Ferrer et al., 2010). MSC ablation has been proven to disrupt hematopoiesis (Raaijmakers et al., 2010). Lymphocytes, originating from HSCs, are a type of white blood cell in the vertebrate immune system. It has recently been shown that MSCs are capable of interacting with lymphocytes. In this review, we discuss current studies on the immunomodulatory properties of MSCs and the effects of lymphocytes on MSCs.

MSCs Target Lymphocytes

Lymphocytes constitute a vital part of the immune system and can be divided into large and small lymphocytes. Large lymphocytes are mainly natural killer cells (NK cells), which are important innate immune cells in the defense against viruses and tumors by cytolysis and secretion of cytokines. Small lymphocytes consist of T-cells and B-cells, which are capable of generating specific immune responses to pathogens, constituting major components of the adaptive immune system. T-cells include CD8+ cytotoxic T-lymphocytes (CTLs) that induce death of target cells or CD4+ helper T-cells (Ths) that regulate other immune cells (Uccelli et al., 2008). Both autologous and allogenic bone marrow MSCs have been found to suppress T-cell proliferation by secreting mediators, including transforming growth factor β1 (TGFβ1) and hepatocyte growth factor (HGF) (Di Nicola et al., 2002). Since then, the immunomodulatory properties of MSCs have attracted extensive attention. It appears that MSCs derived from bone marrow, the orofacial region, and other areas of the body are able to target all subsets of lymphocytes (Fig. 1) (Ren et al., 2008; Zhao et al., 2010).

Figure 1.

Figure 1.

Immunomodulatory properties of mesenchymal stem cells (MSCs). (A) MSCs can target several subsets of lymphocytes, including CD4+ helper T-lymphocytes (Ths), CD8+ cytotoxic T-lymphocytes (CTLs), gammadelta T-cells, natural killer (NK) cells, B-lymphocytes, and regulatory T-lymphocytes (Tregs). These effects may be mediated by several soluble factors secreted by MSCs, including, for example, prostaglandin E2 (PGE2), transforming growth factor-β1 (TGF β1), nitric oxide (NO), indoleamine 2,3-dioxygenase (IDO), or hepatocyte growth factor (HGF). (B) Infused MSCs can induce T-cell apoptosis through FAS/FASL-mediated multiple paracrine interactions and cell-cell contacts, as well as promoting the generation of Tregs, which ultimately leads to immune tolerance. This process consists of the following stages: (1) MSCs use FAS to control monocyte chemotactic protein 1 (MCP-1) secretion, and MCP-1 recruits activated T-cells; (2) MSCs use FASL to induce activated T-cell apoptosis; (3) apoptotic T-cells subsequently trigger macrophages to produce high levels of TGFβ; and (4) the high level of TGFβ up-regulates Tregs to induce immune tolerance.

CD4+ T-helper cells (Ths) play an important role in the adaptive immune system by activating and directing other immune cells through cytokines or a combination of cell/cell interactions (e.g., CD40 and CD40L). It has been shown that MSCs efficiently suppress proliferation of CD4+ Ths by arresting T-cells in the G0/G1 phase (Di Nicola et al., 2002; Glennie et al., 2005; Krampera et al., 2006). Moreover, MSCs are able to reduce Th1-cell-produced interferon γ (IFN-γ) and Th17-cell-produced interleukin-17 (IL17), whereas they enhance Th2 cells to secrete IL-4 (Aggarwal and Pittenger, 2005; Sun et al., 2009). CD8+ CTLs mediate major histocompatibility complex (MHC)-restricted killing of allogenic or virus-infected cells, and they are vital for the graft-vs.-leukemia effect. MSCs have been demonstrated to inhibit CTL formation, thereby down-regulating CTL-mediated cytotoxicity (Rasmusson et al., 2003).

Regulatory T-cells (Tregs) are a functionally distinct CD4+ T-cell population in the peripheral blood. They express the transcription factor forkhead box P3 (FOXP3) to regulate their own development and function to actively suppress autoimmune response (Fontenot et al., 2003). MSCs have been reported to directly or indirectly promote the proliferation of Tregs and enhance their regulatory capacity (Aggarwal and Pittenger, 2005; Maccario et al., 2005; Di Ianni et al., 2008; Selmani et al., 2008). Gammadelta T-cells play an important role in the immunosurveillance of cancer and have been shown to be implicated in acute graft-vs.-host disease (GVHD). MSCs effectively suppress in vitro expansion of gammadelta T-cells without affecting their cytotoxicity (Petrini et al., 2009). MSCs are also potent suppressors of TCRVgamma9Vdelta2(+) gammadelta lymphocyte proliferation, cytokine production, and cytolytic responses in vitro, as mediated by the cyclooxygenase-2 (COX-2)-dependent production of prostaglandin E2 (PGE2) (Martinet et al., 2009). Natural killer (NK) cells are important effector cells of innate immunity and play a key role in antitumor and antiviral effects by their cytotoxic potential and secretion of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and IFN-γ. However, they also contribute to several pathophysiological autoimmune conditions by their cytotoxic activity (Malhotra and Shanker, 2011). MSCs can inhibit the proliferation, cytokine production, and cytotoxic activity of both resting and pre-activated NK cells (Sotiropoulou et al., 2006; Spaggiari et al., 2006).

B-lymphocytes produce antibodies and closely interact with T-cells, thereby contributing to several autoimmune diseases, such as multiple sclerosis. The effects of MSCs on B-cells remain controversial. Most studies have demonstrated that MSCs inhibit B-cell proliferation, differentiation, and antibody secretion in in vitro co-culture assays and in vivo multiple sclerosis models (Augello et al., 2005; Corcione et al., 2006; Gerdoni et al., 2007; Asari et al., 2009). However, other in vitro studies showed that MSCs support B-cell proliferation and stimulate antibody secretion in B-cells (Rasmusson et al., 2007; Traggiai et al., 2008). It is possible that MSC-mediated regulation of B-cells may depend on the developmental stage of B-cells and the local microenvironment.

The immunoregulatory properties of MSCs provide a foundation for the clinical use of MSCs to treat a variety of immune diseases (Table). Since Bartholomew et al. first demonstrated that systemic infusion of allogenic MSCs can prolong skin-graft survival in monkeys by inhibiting T-cells in vivo (Bartholomew et al., 2002), MSC-based immunotherapy has shown successful outcomes in several pre-clinical disease models, such as systemic lupus erythematosus (SLE), rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and systemic sclerosis (SS). In terms of using the systemic infusion of MSCs for patient treatment, Le Blanc et al. first reported that allogenic MSC infusion may provide appropriate therapy for a severe treatment-resistant acute GVHD patient (Le Blanc et al., 2004). Our group and collaborators demonstrated that allogenic MSCs can effectively ameliorate disease activity, improve serologic markers, and reverse renal dysfunction in patients with SLE, through up-regulating Tregs and down-regulating Th17 cells, leading to an immune tolerance (Sun et al., 2009). However, another group showed that autologous MSC infusion failed to ameliorate disease activity in SLE patients (Carrion et al., 2010). These discrepant results may be attributed, at least in part, to the impairment of bone marrow MSCs as observed in SLE patients and SLE-like MRL/lpr mice (Sun et al., 2009). Therefore, it is critical to use healthy MSCs for cell-based immune therapies. MSCs have also shown efficacy and safety in several clinical trials for myocardial infarction, acute lung injury, chronic obstructive pulmonary disease (COPD), diabetes, and Crohn’s disease, a painful inflammatory disease in the bowels and gastrointestinal tract (Boumaza et al., 2009; Hare et al., 2009; Ciccocioppo et al., 2011; Matthay et al., 2010; Mannon, 2011; Mabed and Shahin, 2012). Recently, Osiris Therapeutics, Inc. (Columbia, MD, USA) has received market authorization in Canada to market the first undifferentiated stem cell product, Prochymal®, an intravenously administered formulation of MSCs derived from human bone marrow of healthy adults, for the management of acute GVHD in children who are unresponsive to steroids (Prasad et al., 2011; Pollack, 2012). The emergence of such MSC products provides a promising opportunity for the management of autoimmune diseases by taking advantage of the immunomodulatory properties of MSCs.

Table.

Systemic Infusion of Mesenchymal stem cells (MSCs) for Clinical and Pre-clinical Therapies

Species Disease Major Organs Affected Immunological Mechanisms References
Human Graft vs. host disease (GVHD) Gut and liver Inhibition of donor T-cell reactivity to the normal tissues of the recipient Le Blanc et al., 2004; Prasad et al., 2011
Human and mouse Systemic lupus erythematosus (SLE) Bone and kidney Osteoblastic niche reconstruction; inhibition of Th17 and promotion of Tregs Sun et al., 2009; Yamaza et al., 2010
Human and mouse Systemic sclerosis (SS) Skin Induction of T-cell apoptosis and up-regulation of Tregs through coupling of FAS/ FAS ligand (FASL) Akiyama et al., 2012
Human Crohn’s disease Bowels and gastrointestinal tract Mucosal T-cell apoptosis in the bowels and gastrointestinal tract Ciccocioppo et al., 2011; Mannon, 2011
Human and mouse Chronic obstructive pulmonary disease (COPD), acute lung injury, lung fibrosis Lung Inhibition of pro-inflammatory cytokine production Ortiz et al., 2007; Matthay et al., 2010; Pollack, 2012
Human, rat, and mouse Diabetes Pancreas and renal glomeruli Alteration of T-cell cytokine pattern and preservation of Tregs; inhibition of macrophage infiltration Lee et al., 2006; Boumaza et al., 2009; Mabed and Shahin, 2012
Human, swine, and mouse Myocardial infarction Heart IL-10-mediated switch from infiltration of pro-inflammatory to anti-inflammatory macrophages; SDF-1/CXCR4-induced engraftment Freyman et al., 2006; Hare et al., 2009; Dayan et al., 2011; Dong et al., 2012
Monkey Graft rejection Skin Inhibition of T-cells Bartholomew et al., 2002
Rat Acute renal failure Kidney Inhibition of pro-inflammatory cytokine production Togel et al., 2005
Mouse Colitis Colon and small intestine Induction of T-cell apoptosis and up-regulation of Tregs through coupling of FAS/FASL, or secretion of immunosuppressive factors Zhang et al., 2009; Akiyama et al., 2012
Mouse Encephalomyelitis (EAE) model of multiple sclerosis Nerve system Inhibition of myelin-specific T-cells Zappia et al., 2005
Mouse Rheumatoid arthritis Joint Inhibition of pro-inflammatory cytokine-producing T-cells and induction of Tregs Augello et al., 2007
Mouse Acute pancreatitis Pancreas Reducing infiltration of CD3+ T-cells and up-regulation of Tregs Jung et al., 2011

The promising results of treating immune-related diseases by MSC infusion have led to exploration of the underlying mechanisms. It has been shown that MSCs target lymphocytes through several soluble factors, such as PGE2, TGF-β, nitric oxide (NO), indoleamine 2,3-dioxygenase (IDO), HGF, and human leukocyte antigen G (HLA-G) (Meisel et al., 2004; Sotiropoulou et al., 2006; Sato et al., 2007; Ren et al., 2008, 2009; Selmani et al., 2008; English et al., 2009). Although these studies provided fundamental knowledge for MSC-based immunoregulation, the diverse and conflicting results indicate that the immunomodulatory effects of MSCs are, most likely, a dynamic process, which may be involved in multiple factors. Very recently, our group revealed a new signaling cascade that may contribute to understanding the interplay between infused MSCs and recipient immune cells in MSC-mediated immune therapies. We found that MSCs produce FAS ligand (FASL) to induce activated T-cell apoptosis via cell-cell contact. Additionally, MSCs use FAS, a death receptor known as tumor necrosis factor receptor superfamily member 6, to control secretion of monocyte chemotactic protein 1 (MCP-1), which attracts T-cell migration to ensure cell-cell contact between MSCs and activated T-cells (Akiyama et al., 2012). In a diseased mouse model, including systemic sclerosis (SS) and dextran-sulfate-sodium-induced experimental colitis, apoptotic T-cells, caused by systemic MSC infusion, triggered macrophages to produce high levels of TGFβ, leading to an up-regulation of CD4+CD25+Foxp3+ Tregs. Eventually, up-regulation of Tregs results in an immune tolerance and ameliorates disease phenotype in the animal models (Akiyama et al., 2012). The systemically infused MSCs may also lead to their lodging in the ischemic brain or myocardium, and stromal-cell-derived factor-1 (SDF-1) may play an important role in guiding MSCs to the sites of injury (Li et al., 2005; Dong et al., 2012). Moreover, bone marrow MSCs express C-X-C chemokine receptor type 4 (CXCR-4), the specific receptor of SDF-1, suggesting that the interaction of SDF-1 with CXCR4 may mediate the trafficking of these stem cells to the impaired site (Kortesidis et al., 2005).

The large study-to-study variations in the effects and underlying mechanisms of MSC-lymphocyte interaction may be attributed to the following: (1) variations in MSC properties and status which are due to batch-to-batch variations in MSCs, culture conditions, and human MSC cell division number, etc. (Song et al., 2008; Lee et al., 2012); and (2) variations in in vivo environments derived from different individuals and immune-related diseases (Ren et al., 2008). Rather than only through a single molecule, it is most likely that MSCs exert immunoregulatory effects through a multi-staged biological process involving the use of receptor FAS to control MCP-1 secretion for recruiting activated T-cells and ligand FASL to induce T-cell apoptosis (Akiyama et al., 2012). A full understanding of the mechanisms is the key to solving this problem, and may allow for the use of non-cell-based therapies that replicate the key factors secreted by MSCs.

Importantly, orofacial tissue-derived MSCs have also been demonstrated to have immunoregulatory properties. MSCs from human exfoliated deciduous teeth (SHED) can inhibit secretion of IL-17 in vitro, and they are capable of effectively reversing SLE-associated disorders in MRL/lpr mice by elevating the ratio of Tregs to Th17 cells (Yamaza et al., 2010). Swine MSCs from apical papilla (SCAP) can suppress T-cell proliferation in vitro through an apoptosis-independent mechanism (Ding et al., 2010a). Human MSCs from periodontal ligament (PDLSCs) also possess immunosuppressive properties when co-cultured with activated peripheral blood mononuclear cells (PBMNCs) (Wada et al., 2009). Accumulated evidence shows that gingiva-derived MSCs are a unique and promising cell source for immune therapies (Zhang et al., 2009).

Lymphocytes Affect MSC Survival and Differentiation

Apart from the promising applications in immune therapies, exogenously added MSCs have long been thought capable of generating new bone and associated tissues to replace damaged tissues. The use of culture-expanded MSCs in conjunction with scaffolds has been widely reported for tissue engineering in pre-clinical models and clinical trials. Seed cells, bio-scaffold, and growth factors have long been considered as key factors for tissue engineering, and the majority of studies in this field focus on the development of better bio-scaffolds, especially biocompatible nanomaterials, as well as improvement in the tissue-specific differentiation capabilities of seed cells by exogenous application of growth factors. Despite the marked progress in MSC-based tissue engineering, the main challenge remains the formation of large quantities of high-quality tissue or even complex organs that meet the functional requirements. Recently, it was reported that cells from the recipient microenvironment may participate in cell-based tissue regeneration. Although MSCs are generally considered to be less immunogenic because they constitutively express low levels of MHC class I and are negative for MHC class II (Le Blanc et al., 2003), the survival of implanted MSCs may be affected by the recipient immune system. It has been proved that cytokine-activated NK cells can efficiently lyse both autologous and allogenic MSCs in vitro (Spaggiari et al., 2006; Sotiropoulou et al., 2006). Moreover, T-cells activated by CD3 and CD28 T-cells can induce bone marrow MSC apoptosis through the FAS/FASL pathway (Yamaza et al., 2008). Similarly, activated T-lymphocytes impair orofacial bone/bone-marrow-derived MSCs (OMSCs), suggesting that OMSCs are capable of interacting with systemic immunity (Yamaza et al., 2011). In addition, T-cells induce bone marrow MSC and osteoblast apoptosis through the CD40/CD40L pathway, as observed in some bone-disease-related animal models, such as osteoporosis (Li et al., 2011). Conversely, immune components have been proven to regulate the differentiation of MSCs. For example, it has been shown that pro-inflammatory cytokine TNF-α inhibits MSC adipogenesis and osteoblastogenesis (Suzawa et al., 2003; Lu et al., 2011). These studies suggested that the crosstalk between implanted MSCs and recipient immune cells may play a key role in determining the success of MSC-based tissue regeneration.

Most recently, our group found that host lymphocytes secrete IFN-γ and TNF-α to block MSC-based bone regeneration (Liu et al., 2011). When treating bone marrow MSCs with IFN-γ alone or a combination of IFN-γ and TNF-α in vitro, we found that IFN-γ alone blocks osteogenic differentiation by inducing up-regulation of SMAD family member 6 (SMAD6), thereby inhibiting Runt-related transcription factor 2 (RUNX2), a key transcription factor associated with osteoblast differentiation. In contrast, TNF-α induces MSC apoptosis in a dose-dependent manner. More interestingly, the combination of IFN-γ and TNF-α can accelerate MSC apoptosis through internalization of FAS, with reduction of the anti-apoptotic factors nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB), X-linked inhibitor of apoptosis protein (XIAP), and FLICE-like inhibitory protein (FLIP). By grafting bone marrow MSCs subcutaneously in a mouse model, using hydroxyapatite tricalcium phosphate (HA-TCP) particles as a carrier, we confirmed that recipient T-cells inhibit MSC-based bone regeneration using the same mechanism observed in vitro. Based on this finding, we further applied systemic infusion of Tregs, or local administration of aspirin, to reduce IFN-γ and TNF-α concentration, and found that both methods can markedly alleviate IFN-γ/TNF-α-induced MSC apoptosis, thereby improving MSC-mediated subcutaneous bone formation (Liu et al., 2011). Furthermore, we showed that local aspirin treatment and systemic Treg infusion are able to significantly improve MSC-mediated calvarial bone repair via inhibition of IFN-γ and TNF-α levels (Liu et al., 2011). Therefore, treatment with aspirin or Tregs may provide promising approaches for improving MSC-based tissue engineering (Fig. 2).

Figure 2.

Figure 2.

Recipient T-lymphocytes govern Mesenchymal stem cell (MSC)-based bone regeneration via IFN-γ and TNF-α. Recipient T-cells secrete IFN-γ to inhibit osteogenic differentiation of implanted MSCs by inducing Smad6/Runx2 signaling and TNF-α to trigger apoptotic pathway via activation of caspase 8 and caspase 3. However, a combination of IFN-γ and TNF-α treatment initiates FAS internalization and accelerates caspase 8/3 apoptotic process. As a novel strategy to improve MSC-mediated bone regeneration, local administration of aspirin or systemic infusion of Tregs can reduce the levels of IFN-γ and TNF-α, thereby protecting implanted MSCs from recipient T-cell attack.

MSC Immunomodulatory Properties Contribute to Oral Disease Therapy and Regenerative Dentistry

Immunomodulatory properties of MSCs may play an important role in treating immune-related oral diseases. Among the many immune-related diseases in the orofacial region, bisphosphonate- related osteonecrosis of the jaw (BRONJ) is a critical side-effect of bisphosphonate therapy for metastatic cancer or osteoporosis patients, especially in those who undergo high-dose bisphosphonate and immunosuppressant drug administration. To date, appropriate therapy has not yet been established for the treatment of BRONJ, largely due to a lack of understanding of its patho-physiological mechanisms. We have developed a mouse model of BRONJ-like disease by the administration of zoledronate and dexamethasone, an immunosuppressant drug, and found that such BRONJ-like disease in mice is caused by suppression of Tregs and activation of Th17 cells (Kikuiri et al., 2010). Interestingly, systemic infusion of MSCs is able to prevent and cure BRONJ-like disease, possibly via induction of peripheral tolerance, shown as an inhibition of Th17 cells and elevation of Tregs, thereby supporting the rationale for the use of MSCs as an immunomodulatory approach for BRONJ treatment (Kikuiri et al., 2010). Additionally, local transplantation of bone marrow MSCs with platelet-rich plasma was reported to alleviate BRONJ lesion of a patient undergoing alendronate and pamidronate treatment for osteoporosis, with a complete healing observed in a 30-month follow-up (Cella et al., 2011).

The crosstalk between the locally implanted MSCs and recipient cells also presents implications for cell-based regenerative dentistry, which has attracted extensive attention in the past decade. In a swine model of periodontal defects, autologous and allogenic PDLSC-mediated treatment has been demonstrated to result in a regeneration of PDL and recovery of alveolar bone height (Liu et al., 2008; Ding et al., 2010b). Furthermore, SHED have been proved to engraft and regenerate bone to repair critical-size craniofacial bone defects generated in mouse and swine models (Seo et al., 2008; Zheng et al., 2009). In terms of tooth regeneration, it has been demonstrated that a combination of SCAP and PDLSCs is able to generate a bio-root with periodontal ligament tissues in a swine model (Sonoyama et al., 2006). Because of their capability of forming dentin-pulp-like complexes, DPSCs, SHED, and SCAP have also been demonstrated to lead to dentin/pulp tissue regeneration (Gronthos et al., 2000; Huang et al., 2008). However, the oral cavity is a challenging environment with active immune responses, which potentially affects periodontal, jaw bone, and tooth regeneration. For example, a wide spectrum of micro-organisms is colonized in periodontal regions, and produces a variety of factors that elicit a host response of inflammatory cell recruitment with secretion of pro-inflammatory mediators (Thomas and Puleo, 2011). Such active immune responses may, in turn, hamper the performance of MSC-based tissue engineering in repairing periodontitis-induced alveolar bone defects. Therefore, it is critical to seek anti-inflammatory treatments, for example, by using aspirin or Tregs, to provide promising approaches for improving MSC-based regeneration in orofacial regions.

Emerging evidence shows that dental MSCs are preferable for regenerative dentistry and treating immune-related oral diseases because of the following: (1) Compared with MSCs derived from bone marrow or other sources, dental MSCs present easier accessibility with minimal trauma. Notably, SHED are the first MSCs derived from human exfoliated tissue, a very accessible tissue resource (Miura et al., 2003). (2) Dental MSCs, including SHED, SCAP, PDLSCs and jaw bone MSCs, show a strong immunomodulatory capacity, possibly because of the high frequency of exposure to the inflammatory environment in the oral cavity (Wada et al., 2009; Ding et al., 2010a; Yamaza et al., 2010, 2011). (3) Because of their neural crest origins, dental MSCs show robust multi-potential differentiation capabilities, benefiting the regeneration of orofacial tissues in orofacial context (Chung et al., 2009; Yamaza et al., 2011). (4) Dental MSCs usually present high proliferation rates to provide sufficient numbers of cells for therapy (Gronthos et al., 2000; Miura et al., 2003).

Conclusions and Outlook

Understanding the role of MSCs and their therapeutic potential has led to great strides over the past decade. MSCs were initially considered as having the potential to differentiate into only tissue-specific cells for regenerative medicine; however, they are now appreciated as an essential cell type that possesses important immunomodulatory properties capable of treating a variety of immune-related diseases. MSCs can regulate the intensity of immune response by inducing T-cell apoptosis through the FAS/FASL-pathway, along with multiple paracrine interactions and cell-cell contacts, as well as promote the generation of Tregs, resulting in immune tolerance. Conversely, lymphocytes can inhibit MSC survival and differentiation by secreting the pro-inflammatory cytokines IFN-γ and TNF-α and/or through cell-cell-contact-induced MSC apoptosis, thereby negatively affecting MSC-based tissue regeneration. In light of such an in-depth understanding of the interplay between lymphocytes and MSCs, novel strategies to improve MSC-based tissue engineering have been proposed to reduce IFN-γ and TNF-α levels by local aspirin treatment or the systemic infusion of Tregs.

Given the complexity of immune profiles in different individuals and various immune-related diseases, the final outcome of the interplay between lymphocytes and MSCs is very likely to be significantly influenced by the in vivo microenvironments. Therefore, it is important to further characterize differences in the immunomodulatory performances of infused MSCs in individuals with different diseases. Conversely, it will be necessary to further understand how the host lymphocytes affect MSCs in various scenarios, such as during tissue injury and immune diseases. Moreover, it will be interesting to clarify whether the locally implanted MSCs can contribute to both tissue repair and immunomodulatory response, thereby preventing potential autoimmune reactions by the host immune system at the site of injury. Through better understanding of the mechanisms underlying the interplay between lymphocytes and MSCs under different physiological and pathological conditions, we will be able to develop promising strategies to improve regenerative medicine and the treatment of immune-mediated diseases.

Footnotes

Some studies reported in this manuscript were supported by grants from the National Institute of Dental and Craniofacial Research, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, USA (R01 DE017449 and R01 DE019932 to S.S).

The authors declare no potential conflicts of interest with respect to the authorship and/or publication of this article.

References

  1. Aggarwal S, Pittenger MF. (2005). Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105:1815-1822. [DOI] [PubMed] [Google Scholar]
  2. Akiyama K, Chen C, Wang D, Xu X, Qu C, Yamaza T, et al. (2012). Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis. Cell Stem Cell 10:544-555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Asari S, Itakura S, Ferreri K, Liu CP, Kuroda Y, Kandeel F, et al. (2009). Mesenchymal stem cells suppress B-cell terminal differentiation. Exp Hematol 37:604-615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Augello A, Tasso R, Negrini SM, Amateis A, Indiveri F, Cancedda R, et al. (2005). Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur J Immunol 35:1482-1490. [DOI] [PubMed] [Google Scholar]
  5. Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. (2007). Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum 56:1175-1186. [DOI] [PubMed] [Google Scholar]
  6. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, et al. (2002). Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 30:42-48. [DOI] [PubMed] [Google Scholar]
  7. Bi Y, Ehirchiou D, Kilts TM, Inkson CA, Embree MC, Sonoyama W, et al. (2007). Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nat Med 13:1219-1227. [DOI] [PubMed] [Google Scholar]
  8. Boumaza I, Srinivasan S, Witt WT, Feghali-Bostwick C, Dai Y, Garcia-Ocana A, et al. (2009). Autologous bone marrow-derived rat mesenchymal stem cells promote PDX-1 and insulin expression in the islets, alter T cell cytokine pattern and preserve regulatory T cells in the periphery and induce sustained normoglycemia. J Autoimmun 32:33-42. [DOI] [PubMed] [Google Scholar]
  9. Caplan AI. (1991). Mesenchymal stem cells. J Orthop Res 9:641-650. [DOI] [PubMed] [Google Scholar]
  10. Carrion F, Nova E, Ruiz C, Diaz F, Inostroza C, Rojo D, et al. (2010). Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus 19:317-322. [DOI] [PubMed] [Google Scholar]
  11. Cella L, Oppici A, Arbasi M, Moretto M, Piepoli M, Vallisa D, et al. (2011). Autologous bone marrow stem cell intralesional transplantation repairing bisphosphonate related osteonecrosis of the jaw. Head Face Med 7:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chung IH, Yamaza T, Zhao H, Choung PH, Shi S, Chai Y. (2009). Stem cell property of postmigratory cranial crest cells and their utility in alveolar bone regeneration and tooth development. Stem Cells 27:866-877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Ciccocioppo R, Bernardo ME, Sgarella A, Maccario R, Avanzini MA, Ubezio C, et al. (2011). Autologous bone marrow-derived mesenchymal stromal cells in the treatment of fistulising Crohn’s disease. Gut 60:788-798. [DOI] [PubMed] [Google Scholar]
  14. Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, et al. (2006). Human mesenchymal stem cells modulate B-cell functions. Blood 107:367-372. [DOI] [PubMed] [Google Scholar]
  15. da Silva Meirelles L, Chagastelles PC, Nardi NB. (2006). Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 119(Pt 11):2204-2213. [DOI] [PubMed] [Google Scholar]
  16. Dayan V, Yannarelli G, Billia F, Filomeno P, Wang XH, Davies JE, et al. (2011). Mesenchymal stromal cells mediate a switch to alternatively activated monocytes/macrophages after acute myocardial infarction. Basic Res Cardiol 106:1299-1310. [DOI] [PubMed] [Google Scholar]
  17. Di Ianni M, Del Papa B, De Ioanni M, Moretti L, Bonifacio E, Cecchini D, et al. (2008). Mesenchymal cells recruit and regulate T regulatory cells. Exp Hematol 36:309-318. [DOI] [PubMed] [Google Scholar]
  18. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al. (2002). Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 99:3838-3843. [DOI] [PubMed] [Google Scholar]
  19. Ding G, Liu Y, An Y, Zhang C, Shi S, Wang W, et al. (2010a). Suppression of T cell proliferation by root apical papilla stem cells in vitro. Cells Tissues Organs 191:357-364. [DOI] [PubMed] [Google Scholar]
  20. Ding G, Liu Y, Wang W, Wei F, Liu D, Fan Z, et al. (2010b). Allogeneic periodontal ligament stem cell therapy for periodontitis in swine. Stem Cells 28:1829-1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Dong F, Harvey J, Finan A, Weber K, Agarwal U, Penn MS. (2012). Myocardial CXCR4 expression is required for mesenchymal stem cell mediated repair following acute myocardial infarction. Circulation 126:314-324. [DOI] [PubMed] [Google Scholar]
  22. English K, Ryan JM, Tobin L, Murphy MJ, Barry FP, Mahon BP. (2009). Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4+CD25(High) forkhead box P3+ regulatory T cells. Clin Exp Immunol 156:149-160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Fontenot JD, Gavin MA, Rudensky AY. (2003). Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4:330-336. [DOI] [PubMed] [Google Scholar]
  24. Freyman T, Polin G, Osman H, Crary J, Lu M, Cheng L, et al. (2006). A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction. Eur Heart J 27:1114-1122. [DOI] [PubMed] [Google Scholar]
  25. Friedenstein AJ, Chailakhjan RK, Lalykina KS. (1970). The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 3:393-403. [DOI] [PubMed] [Google Scholar]
  26. Gerdoni E, Gallo B, Casazza S, Musio S, Bonanni I, Pedemonte E, et al. (2007). Mesenchymal stem cells effectively modulate pathogenic immune response in experimental autoimmune encephalomyelitis. Ann Neurol 61:219-227. [DOI] [PubMed] [Google Scholar]
  27. Glennie S, Soeiro I, Dyson PJ, Lam EW, Dazzi F. (2005). Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 105:2821-2827. [DOI] [PubMed] [Google Scholar]
  28. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. (2000). Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA 97:13625-13630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, et al. (2009). A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol 54:2277-2286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Huang GT, Sonoyama W, Liu Y, Liu H, Wang S, Shi S. (2008). The hidden treasure in apical papilla: the potential role in pulp/dentin regeneration and bioroot engineering. J Endod 34:645-651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Jung KH, Song SU, Yi T, Jeon MS, Hong SW, Zheng HM, et al. (2011). Human bone marrow-derived clonal mesenchymal stem cells inhibit inflammation and reduce acute pancreatitis in rats. Gastroenterology 140:998-1008. [DOI] [PubMed] [Google Scholar]
  32. Kikuiri T, Kim I, Yamaza T, Akiyama K, Zhang Q, Li Y, et al. (2010). Cell-based immunotherapy with mesenchymal stem cells cures bisphosphonate-related osteonecrosis of the jaw-like disease in mice. J Bone Miner Res 25:1668-1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Kortesidis A, Zannettino A, Isenmann S, Shi S, Lapidot T, Gronthos S. (2005). Stromal-derived factor-1 promotes the growth, survival, and development of human bone marrow stromal stem cells. Blood 105:3793-3801. [DOI] [PubMed] [Google Scholar]
  34. Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, et al. (2006). Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 24:386-398. [DOI] [PubMed] [Google Scholar]
  35. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. (2003). HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 31:890-896. [DOI] [PubMed] [Google Scholar]
  36. Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, et al. (2004). Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 363:1439-1441. [DOI] [PubMed] [Google Scholar]
  37. Lee EJ, Choi EK, Kang SK, Kim GH, Park JY, Kang HJ, et al. (2012). N-cadherin determines individual variations in the therapeutic efficacy of human umbilical cord blood-derived mesenchymal stem cells in a rat model of myocardial infarction. Mol Ther 20:155-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Lee RH, Seo MJ, Reger RL, Spees JL, Pulin AA, Olson SD, et al. (2006). Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc Natl Acad Sci USA 103:17438-17443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Li JY, Tawfeek H, Bedi B, Yang X, Adams J, Gao KY, et al. (2011). Ovariectomy disregulates osteoblast and osteoclast formation through the T-cell receptor CD40 ligand. Proc Natl Acad Sci USA 108:768-773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Li Y, Chen J, Zhang CL, Wang L, Lu D, Katakowski M, et al. (2005). Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells. Glia 49:407-417. [DOI] [PubMed] [Google Scholar]
  41. Liu Y, Zheng Y, Ding G, Fang DJ, Zhang CM, Bartold M, et al. (2008). Periodontal ligament stem cell-mediated treatment for periodontitis in miniature swine. Stem Cells 26:1065-1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Liu Y, Wang L, Kikuiri T, Akiyama K, Chen C, Xu X, et al. (2011). Mesenchymal stem cell-based tissue regeneration is governed by recipient T lymphocytes via IFN-gamma and TNF-alpha. Nat Med 17:1594-1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Lu X, Beck GR, Jr., Gilbert LC, Camalier CE, Bateman NW, Hood BL, et al. (2011). Identification of the homeobox protein Prx1 (MHox, Prrx-1) as a regulator of osterix expression and mediator of tumor necrosis factor alpha action in osteoblast differentiation. J Bone Miner Res 26:209-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Mabed M, Shahin M. (2012). Mesenchymal stem cell-based therapy for the treatment of type 1 diabetes mellitus. Curr Stem Cell Res Ther 7:179-190. [DOI] [PubMed] [Google Scholar]
  45. Maccario R, Podesta M, Moretta A, Cometa A, Comoli P, Montagna D, et al. (2005). Interaction of human mesenchymal stem cells with cells involved in alloantigen-specific immune response favors the differentiation of CD4+ T-cell subsets expressing a regulatory/suppressive phenotype. Haematologica 90:516-525. [PubMed] [Google Scholar]
  46. Malhotra A, Shanker A. (2011). NK cells: immune cross-talk and therapeutic implications. Immunotherapy 3:1143-1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Mannon PJ. (2011). Remestemcel-L: human mesenchymal stem cells as an emerging therapy for Crohn’s disease. Expert Opin Biol Ther 11:1249-1256. [DOI] [PubMed] [Google Scholar]
  48. Martinet L, Fleury-Cappellesso S, Gadelorge M, Dietrich G, Bourin P, Fournie JJ, et al. (2009). A regulatory cross-talk between Vgamma9Vdelta2 T lymphocytes and mesenchymal stem cells. Eur J Immunol 39:752-762. [DOI] [PubMed] [Google Scholar]
  49. Matthay MA, Thompson BT, Read EJ, McKenna DH, Liu KD, Calfee CS, et al. (2010). Therapeutic potential of mesenchymal stem cells for severe acute lung injury. Chest 138:965-972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Meisel R, Zibert A, Laryea M, Gobel U, Daubener W, Dilloo D. (2004). Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood 103:4619-4621. [DOI] [PubMed] [Google Scholar]
  51. Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, et al. (2010). Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466:829-834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, et al. (2003). SHED: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci USA 100:5807-5812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Morsczeck C, Gotz W, Schierholz J, Zeilhofer F, Kuhn U, Mohl C, et al. (2005). Isolation of precursor cells (PCs) from human dental follicle of wisdom teeth. Matrix Biol 24:155-165. [DOI] [PubMed] [Google Scholar]
  54. Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, Go K, et al. (2007). Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci USA 104:11002-11007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Petrini I, Pacini S, Petrini M, Fazzi R, Trombi L, Galimberti S. (2009). Mesenchymal cells inhibit expansion but not cytotoxicity exerted by gamma-delta T cells. Eur J Clin Invest 39:813-818. [DOI] [PubMed] [Google Scholar]
  56. Pollack A. (2012). A stem-cell-based drug gets approval in Canada. New York Times Online. URL accessed on 8/15/2012 at: http://www.nytimes.com/2012/05/18/health/a-stem-cell-based-drug-gets-approval-in-canada.html.
  57. Prasad VK, Lucas KG, Kleiner GI, Talano JA, Jacobsohn D, Broadwater G, et al. (2011). Efficacy and safety of ex vivo cultured adult human mesenchymal stem cells (Prochymal™) in pediatric patients with severe refractory acute graft-versus-host disease in a compassionate use study. Biol Blood Marrow Transplant 17:534-541. [DOI] [PubMed] [Google Scholar]
  58. Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. (2010). Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 464:852-857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Rasmusson I, Ringden O, Sundberg B, Le Blanc K. (2003). Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation 76:1208-1213. [DOI] [PubMed] [Google Scholar]
  60. Rasmusson I, Le Blanc K, Sundberg B, Ringden O. (2007). Mesenchymal stem cells stimulate antibody secretion in human B cells. Scand J Immunol 65:336-343. [DOI] [PubMed] [Google Scholar]
  61. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. (2008). Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell 2:141-150. [DOI] [PubMed] [Google Scholar]
  62. Ren G, Su J, Zhang L, Zhao X, Ling W, L’Huillie A, et al. (2009). Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells 27:1954-1962. [DOI] [PubMed] [Google Scholar]
  63. Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S, Saggio I, et al. (2007). Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 131:324-336. [DOI] [PubMed] [Google Scholar]
  64. Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, Nagai T, et al. (2007). Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 109:228-234. [DOI] [PubMed] [Google Scholar]
  65. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. (2008). Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells 26:212-222. [DOI] [PubMed] [Google Scholar]
  66. Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, et al. (2004). Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 364:149-155. [DOI] [PubMed] [Google Scholar]
  67. Seo BM, Sonoyama W, Yamaza T, Coppe C, Kikuiri T, Akiyama K, et al. (2008). SHED repair critical-size calvarial defects in mice. Oral Dis 14:428-434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Shi S, Gronthos S. (2003). Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res 18:696-704. [DOI] [PubMed] [Google Scholar]
  69. Song SU, Kim CS, Yoon SP, Kim SK, Lee MH, Kang JS, et al. (2008). Variations of clonal marrow stem cell lines established from human bone marrow in surface epitopes, differentiation potential, gene expression, and cytokine secretion. Stem Cells Dev 17:451-461. [DOI] [PubMed] [Google Scholar]
  70. Sonoyama W, Liu Y, Fang D, Yamaza T, Seo BM, Zhang C, et al. (2006). Mesenchymal stem cell-mediated functional tooth regeneration in swine. PLoS One 1:e79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN, Papamichail M. (2006). Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells 24:74-85. [DOI] [PubMed] [Google Scholar]
  72. Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. (2006). Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood 107:1484-1490. [DOI] [PubMed] [Google Scholar]
  73. Sun L, Akiyama K, Zhang H, Yamaza T, Hou Y, Zhao S, et al. (2009). Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells 27:1421-1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Suzawa M, Takada I, Yanagisawa J, Ohtake F, Ogawa S, Yamauchi T, et al. (2003). Cytokines suppress adipogenesis and PPAR-gamma function through the TAK1/TAB1/NIK cascade. Nat Cell Biol 5:224-230. [DOI] [PubMed] [Google Scholar]
  75. Thomas MV, Puleo DA. (2011). Infection, inflammation, and bone regeneration: a paradoxical relationship. J Dent Res 90:1052-1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Togel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C. (2005). Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol 289:F31-F42. [DOI] [PubMed] [Google Scholar]
  77. Traggiai E, Volpi S, Schena F, Gattorno M, Ferlito F, Moretta L, et al. (2008). Bone marrow-derived mesenchymal stem cells induce both polyclonal expansion and differentiation of B cells isolated from healthy donors and systemic lupus erythematosus patients. Stem Cells 26:562-569. [DOI] [PubMed] [Google Scholar]
  78. Uccelli A, Moretta L, Pistoia V. (2008). Mesenchymal stem cells in health and disease. Nat Rev Immunol 8:726-736. [DOI] [PubMed] [Google Scholar]
  79. Wada N, Menicanin D, Shi S, Bartold PM, Gronthos S. (2009). Immunomodulatory properties of human periodontal ligament stem cells. J Cell Physiol 219:667-676. [DOI] [PubMed] [Google Scholar]
  80. Yamaza T, Miura Y, Bi Y, Liu Y, Akiyama K, Sonoyama W, et al. (2008). Pharmacologic stem cell based intervention as a new approach to osteoporosis treatment in rodents. PLoS One 3:e2615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Yamaza T, Kentaro A, Chen C, Liu Y, Shi Y, Gronthos S, et al. (2010). Immunomodulatory properties of stem cells from human exfoliated deciduous teeth. Stem Cell Res Ther 1:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Yamaza T, Ren G, Akiyama K, Chen C, Shi Y, Shi S. (2011). Mouse mandible contains distinctive mesenchymal stem cells. J Dent Res 90:317-324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E, et al. (2005). Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood 106:1755-1761. [DOI] [PubMed] [Google Scholar]
  84. Zhang Q, Shi S, Liu Y, Uyanne J, Shi Y, Le AD. (2009). Mesenchymal stem cells derived from human gingiva are capable of immunomodulatory functions and ameliorate inflammation-related tissue destruction in experimental colitis. J Immunol 183:7787-7798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Zhao S, Wehner R, Bornhauser M, Wassmuth R, Bachmann M, Schmitz M. (2010). Immunomodulatory properties of mesenchymal stromal cells and their therapeutic consequences for immune-mediated disorders. Stem Cells Dev 19:607-614. [DOI] [PubMed] [Google Scholar]
  86. Zheng Y, Liu Y, Zhang CM, Zhang HY, Li WH, Shi S, et al. (2009). Stem cells from deciduous tooth repair mandibular defect in swine. J Dent Res 88:249-254. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Dental Research are provided here courtesy of International and American Associations for Dental Research

RESOURCES