Skip to main content
Journal of Cellular and Molecular Medicine logoLink to Journal of Cellular and Molecular Medicine
. 2015 Jun 8;19(9):2067–2074. doi: 10.1111/jcmm.12613

Do oral bacteria alter the regenerative potential of stem cells? A concise review

Kyriaki Chatzivasileiou a,*, Katja Kriebel a, Gustav Steinhoff c, Bernd Kreikemeyer b, Hermann Lang a
PMCID: PMC4568911  PMID: 26058313

Abstract

Mesenchymal stem cells (MSCs) are widely recognized as critical players in tissue regeneration. New insights into stem cell biology provide evidence that MSCs may also contribute to host defence and inflammation. In case of tissue injury or inflammatory diseases, e.g. periodontitis, stem cells are mobilized towards the site of damage, thus coming in close proximity to bacteria and bacterial components. Specifically, in the oral cavity, complex ecosystems of commensal bacteria live in a mutually beneficial state with the host. However, the formation of polymicrobial biofilm communities with pathogenic properties may trigger an inadequate host inflammatory-immune response, leading to the disruption of tissue homoeostasis and development of disease. Because of their unique characteristics, MSCs are suggested as crucial regulators of tissue regeneration even under such harsh environmental conditions. The heterogeneous effects of bacteria on MSCs across studies imply the complexity underlying the interactions between stem cells and bacteria. Hence, a better understanding of stem cell behaviour at sites of inflammation appears to be a key strategy in developing new approaches for in situ tissue regeneration. Here, we review the literature on the effects of oral bacteria on cell proliferation, differentiation capacity and immunomodulation of dental-derived MSCs.

Keywords: stem cells, bacteria, regeneration, inflammation, periodontitis

Introduction

Mesenchymal stem cells (MSCs), also referred to as multipotent mesenchymal stromal cells, are present in all the organs throughout the body and play a key role in tissue regeneration. Aside from their ability to orchestrate regeneration processes, MSCs are newly proposed as critical players in host defence and inflammation 1.

Under physiological conditions, the oral cavity, gastrointestinal tract and skin home complex ecosystems of commensal bacteria that live in a mutually beneficial state with the host. In case of tissue injury or inflammatory diseases, stem cells are mobilized towards the site of damage, thus coming to close vicinity of bacteria and bacterial components. Bacterial infection of stem cells could even lead to long-term functional consequences for the host 2. Recent reports suggest that MSCs may be able to actively participate in the control of infectious challenges by direct targeting of bacteria and through indirect effects on the host primary and adaptive immune response 3.

Oral microflora

The oral cavity, like the skin, the respiratory tract and the gut, is habitat to a plethora of microbiota living in symbiosis with the host 4. The oral microflora is known to contain over 700 species of aerobic and anaerobic bacteria 5. These organisms can be isolated from tooth surfaces, periodontal pockets and other oral sites such as the tongue and oral mucous membranes 6. Oral microbiota grow as complex, mixed, interdependent colonies organized in biofilms 7. Reports in the literature suggest that oral bacterial biofilms may contain more than 105 microorganisms 8, while the concentrations and compositions of pathogenic bacteria in the subgingival biofilm vary greatly depending on the local microenvironmental conditions 9,10. The bacterial genera which are mostly represented in the oral cavity include the following: Gemella, Granulicatella, Streptococcus, Veillonella, Neisseria, Haemophilus, Rothia, Actinomyces, Prevotella, Capnocytophaga, Porphyromona, Fusobacterium, Corynebacterium, Cardiobacterium, Campylobacter, Corynebacterium, Atopobium and Bergeyella 1113. It should be noticed that almost 60% of the species detected by new molecular methods are not presently cultivable and remain uncharacterized 11.

The natural oral microflora is vital for the normal development and physiological integrity of the oral cavity. It also contributes to host defence by excluding exogenous microorganisms 14. It is widely recognized that the maintenance of an ecologically balanced biodiversity of the microflora within the oral cavity is crucial not only to the oral health but also to the general health of the host 15. Microbes have commensal relationships with their co-habitants, while being symbiotic with their host 16. However, ecological shifts may lead to pathological conditions, which alter the relationships between microbes and the host 17. In disease, pathogenic bacteria grow with disregard to their co-habitant bacteria and express their virulence properties, so that the host becomes infected or susceptible to infection 16.

Periodontitis

Periodontitis is a bacterially induced inflammatory disease of the supporting tissues of the teeth. It represents one of the major dental diseases that affect human populations worldwide at high prevalence rates and has a huge economic impact on national health care systems 18. In fact, periodontitis is characterized by progressive periodontal tissue destruction that may finally lead to the loosening and subsequent loss of teeth 19. The predominant pathogens involved in periodontitis are Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, Prevotella intermedia, Fusobacterium nucleatum, Tannerella forsythia, and Eikenella corrodens, and Treponema denticola 15.In addition, several forms of uncultivable spirochetes are supposed to play a major role in the pathogenesis of this disease 20.

Periodontal pathogens induce tissue destruction by activating the host defence. The infection of periodontal tissues is accompanied by the release of bacterial leucotoxins, collagenases, fibrinolysins and other proteases that break down host tissues and may result in gingival inflammation 21. Specifically, microbial components, like lipopolysaccharide (LPS), have the capacity to activate macrophages and lymphocytes to synthesize and secrete a wide array of molecules including cytokines, prostaglandins, hydrolytic enzymes and tumour necrosis factor alpha, which in turn stimulate the effectors of periodontal tissue breakdown 22. Cell activation occurs mainly through two members of the Toll-like receptor (TLR) family, TLR2 and TLR4 that are documented as predominant signalling receptors for most bacterial components 23,24.

Once a periodontal pocket forms and becomes colonized by bacteria, the pathologic situation becomes irreversible 18. The conventional periodontal treatment involves the mechanical removal of the pathogenic dental biofilm. Successful clinical outcomes such as probing depth reduction and gain of clinical attachment after treatment are well documented in a plethora of studies 2527. However, histological analyses of healed periodontal tissues reveal in most of the cases the presence of an epithelial lining along the treated root surfaces of the teeth, instead of true periodontal regeneration 28.

Dental stem cells

Stem cells are defined by their capacity to self-renew and differentiate into multiple cell lineages. One of the most studied adult stem cell types are MSCs 29. Friedenstein et al. first described bone marrow stem cells (BMSCs) as a heterogeneous population of multipotent cells derived from bone marrow aspirates with the ability to adhere to plastic surfaces and form colonies of fibroblast-like cells within the first days of cultivation 30,31. Although MSCs were originally isolated from the bone marrow, similar populations of mesenchymal precursors were isolated from other tissues, including adipose tissue 32, amniotic fluid 33, foetal liver 34 and umbilical cord blood (UCB) 35.

During the last decades, rapid progress in dental research has shed light on the molecular and cellular biology of periodontal tissue development. Recently, multipotent cells have been successfully isolated from several dental tissues including dental pulp 36, dental follicle 37, exfoliated deciduous teeth 38 and the root apical papilla 39. Several in vitro and in vivo studies on dental stem cells (DSCs) provide evidence of their multipotent character and their key role in periodontal regeneration 40. It has been demonstrated that DSCs have a fibroblast-like morphology and are plastic-adherent. Similar to other stem cell populations, DSCs express several surface markers such as CD10, CD13, CD29, CD44, CD53, CD59, CD73, CD90 and CD105, and do not express CD34, CD45 or HLA-DR 41,42. Demonstration of self-renewal ability and multilineage differentiation capacity are additional indications of their stem cell phenotype. Indeed, it has been proven that DSCs are able to form single cell-derived colonies and differentiate into several lineages, when induced by special media in vitro 43.

Clinical relevance

The identification of DSCs has stimulated interest in the potential use of cell-based therapies as prospective alternatives to existing therapeutic approaches for the repair and regeneration of the periodontium 44. One of the critical requirements for the success of such therapeutic interventions would be the repopulation of the periodontal wound by ex vivo expanded progenitor populations or the mobilization of endogenous progenitor cells capable of promoting regeneration 45. Specifically, DSCs grafts may support the restoration of the complex ultrastructure of the periodontal ligament and the dynamic functional relationships of its components. Numerous animal studies have already proved the regenerative potency of these cell populations in vivo 46.

However, one of the growing concerns in dental research is the exposure of DSCs to the inflamed microenvironment of periodontal pockets 47. This may affect many cell properties such as self-renewal, differentiation potential, production of cytokines and extracellular matrix compounds secretion. Sorrell and Caplan demonstrated that multipotent cell grafts might trigger regenerative processes not only through direct commitment, but also by infiltrating inflammatory or antigen-presenting cells 48. Such a regenerative microenvironment may impel self-regulated regenerative cascades and limit the area of damage in the inflamed adult tissues 49. Hence, a better understanding of cell behaviour at sites of bacterial infection appears to be a key strategy for the development of new approaches for periodontal regeneration.

In vitro experimental models

The microenvironment of a periodontal pocket is characterized by the constant presence of bacterial biofilms. This condition results in a continuous cross-talk of periodontal tissue cells with a wide variety of oral microorganisms. Further, in periodontitis, several types of host immune cells (e.g. neutrophils and macrophages) migrate to the site of inflammation 50. The better understanding of the complex cell–bacteria interactions is essential for the development of successful periodontal therapies. While several in vivo models have been already used, the design of an in vitro model that could sufficiently mimic the in vivo situation of inflamed periodontal tissues remains to be developed 51.

Till now most of the in vitro experimental settings are based on the analysis of the LPS effects on cells. Lipopolysaccharide is a major membrane component of Gram-negative bacteria and can be derived from several bacterial species, e.g. Escherichia coli or P. gingivalis 47,52,53. The easy isolation method and the fact that LPS is responsible for many of the inflammatory responses and pathogenic effects of Gram-negative bacteria are the main arguments for the use of LPS in numerous in vitro experiments. Experimental settings using heat-inactivated or sonicated bacteria have also been proposed as models that may correspond to the in vivo condition of bacterial infection 54,55. Further methods used for the analysis of cell–bacteria interactions are based on the fact that periopathogenic bacterial pathogens produce a broad array of potential virulence factors apart from LPS that are released into the gingival crevicular fluid 56. Thus, the culture of cells with bacterial pre-conditioned medium or the co-cultivation of cells and bacteria in transwell systems have been used to evaluate the secretion of soluble factors and the activation of cellular downstream cascades by bacteria 57,58. Although many biological effects can be elicited by non-viable bacteria, it is known that some cell responses require the presence of live bacteria 59.

Experimental models utilizing microorganisms in a planktonic state were used to imitate the periodontal infection 60. Nevertheless, such systems may not adequately portray the bacterial challenge conferred by a polymicrobial, biofilm-induced disease, such as periodontitis 61. Thus, in vitro multispecies dental biofilm settings have been proposed as laboratory models that better mimic the environment of chronic periodontitis 6264. Finally, cell invasion is a common strategy of pathogens that facilitates their escape from host immune system, access to nutrients, persistence and spread into tissues 65. Recent studies using viable bacteria have been demonstrated as models for the analysis of host cell invasion processes such as bacterial adherence and internalization by cells 66,67. However, the subgingival bacteria that are closely correlated with periodontitis are mainly anaerobes. The co-culture of these bacteria with oxygen-requiring cells in conventional systems is not possible 68. Therefore, one weak point of the experimental studies on periodontal infection is the fact that most in vitro settings are conducted under aerated conditions. Given the fact that aerotolerance of strictly anaerobic pathogens like P. gingivalis is very low, the interpretation of such experimental results may not directly reflect the in vivo situation 69. Until now only few models have been proposed utilizing direct contact between live obligate anaerobic bacteria and human cell lines under oxygen-free conditions 70,71.

Influence of oral bacteria on stem cells

Effects on cell viability and proliferation of stem cells

Cell proliferation is fundamental in tissue homoeostasis and can be controlled by either physiological or pathological conditions. Previous studies have demonstrated that LPS derived from periopathogenic bacteria may induce controversial effects on the proliferation of periodontal ligament fibroblasts 7274. Currently, the possible effect of bacteria on the proliferative rates of multipotent cells is in the focus of interest of several research groups. Kato et al. demonstrated that P. gingivalis LPS promoted cell proliferation in periodontal ligament stem cells (PDLSCs) 53. Stimulation of TLR2 also led to enhanced proliferation of adult BMSCs 75. Further, Jiang et al. and Buchon et al. proposed that intestinal stem cells are able to maintain tissue homoeostasis by increasing their proliferation rates to repair tissue damage at sites of infection through the JAK-STAT signalling pathway 76,77.

On the contrary, according to an in vitro study on canine adipose-derived MSCs (ADSCs), gastrointestinal microbes did not induce cell death nor diminished cell proliferation. Previous studies on dental follicle progenitors also demonstrated that cell viability of both dental follicle progenitor cells (DFPCs) and BMSCs was not affected by P. gingivalis LPS treatment 47,78. In addition, TLR ligands such as LPS and flagellin do not alter proliferation rates of a newly identified population of pluripotent UCB cells, which are termed as unrestricted somatic stem cells 79. Nevertheless, LPS and extracts from Streptococcus mutans treatment were able to inhibit the proliferation of dental pulp stem cells (DPSCs) in vitro 80.

These heterogeneous effects of bacteria on the induction or inhibition of cell proliferation across studies could imply the complexity of the underlying mechanisms that rule the interactions between host cells and bacteria. Specifically, cell response to bacterial stimuli seems to be associated with the cell type, bacterial strain and specific bacterial components used in each experimental setting 81.

Effects on differentiation capacity of stem cells

The ability of stem cells to differentiate into multiple lineages is well documented. Especially the differentiation capacity of DSCs across the osteogenic, chondrogenic, adipogenic and neurogenic lineages has been demonstrated from several research groups in the last years 82,83. Nevertheless, the impact of bacteria on the differentiation capacity of stem cells remains to be explored. In a recent study, Ronay et al. demonstrated that infected periodontal granulation tissues harbour cells expressing embryonic stem cell markers, and exhibit osteogenic capacities 84. These results are in accordance with other studies demonstrating elevated alkaline phosphatase (ALP) activity, an early marker for osteogenic differentiation, and calcium deposition after E. coli LPS treatment of BMSCs 52.

Nevertheless, an increased ALP activity after LPS treatment may not always lead to formation of mineralized nodules in vitro. It is suggested that LPS may partly block the progression of molecular processes involved in osteogenic differentiation 47. Interestingly, Abe et al. demonstrated that low concentrations of P. gingivalis extracts improve the osteogenic differentiation of human dental pulp-derived cells while high concentrations may inhibit ALP activity and bone sialoprotein gene expression 85. High concentration of sodium butyrate, a major metabolic by-product of anaerobic Gram-negative periodontopathogenic bacteria, could inhibit the osteoblastic differentiation and mineralized nodule formation in an osteoblastic cell line in vitro 86. In accordance with these results P. gingivalis LPS was shown to suppress the osteoblastic differentiation in both PDLSCs and DFPCs 47,53. Nomiyama et al. suggested that Gram-negative bacterial infection might down-regulate the odontoblastic properties of rat pulp progenitor cells after stimulation with A. actinomycetemcomitans LPS 87. Treatment with LPS from P. gingivalis was shown to impair both ALP activity and the formation of mineral deposits in DPSCs 88. In this context, TLR ligands have been proposed as possible regulators of stem cell differentiation state in vitro 78.

Further, P. gingivalis fimbriae are proposed as potent inducers of a monocyte/macrophage tumour cell line differentiation, via cyclic nucleotide-independent protein kinase C 89. However, P. gingivalis fimbriae were proved unable to alter the osteoblastic differentiation and mineralization in long-term mouse calvarial osteoblast cultures 90.

Effects on the immunomodulatory properties of stem cells

In the last years the immunomodulatory functions of the stem cells have been in the focus of research 91. Accumulating evidence indicated that MSCs may affect neighbouring innate and adaptive immune cells by two main ways: the direct cell–cell contact and the release of a variety of soluble factors 9297. Gingiva-derived MSCs (GMSCs) were shown to have immunomodulatory functions. Specifically, GMSCs were able to suppress peripheral blood lymphocyte proliferation and induce expression of a wide panel of immunosuppressive factors including interleukin (IL)-10, indoleamine 2,3-dioxygenase, inducible nitric oxide synthase and cyclooxygenase 2 in response to the inflammatory cytokine, interferon-γ 98. However, the behaviour of cells under the direct influence of bacteria remains less understood.

Reed et al. recently demonstrated that human embryonic stem cell-derived endothelial cells (hESC-ECs) are TLR4 deficient but respond to bacteria via the intracellular receptor nucleotide-binding oligomerization domain-containing protein 1 (NOD1). The authors suggested that hESC-ECs may be protected from unwanted TLR4-mediated vascular inflammation, thus offering a potential therapeutic advantage 99. On the other side, studies on DFPCs revealed the expression of TLR2 and TLR4 in both mRNA and protein level. Nevertheless, when these cells were treated with P. gingivalis LPS no effect on the expression of pro-inflammatory cytokines has been observed 47,78. Further, treatment with TLR4 agonist augmented the suppressive potential of DFPCs and increased the transforming growth factor-beta production 100. In accordance with these results canine ADSCs were shown to enhance immunomodulation after interacting with gastrointestinal microbes in vitro 101.

It is reported that LPS is able to induce the expression of the nuclear factor κB (NF-κB) -dependent gene IL-8 by DPSCs 102. He et al. suggested that LPS-mediated transcriptional and post-translational up-regulation of IL-8 in DPSCs is a process that also involves TLR4, myeloid differentiation primary response gene 88 (MyD88), NF-κB and mitogen-activated protein kinases 103. Further, Mei et al. demonstrated that MSCs improve the survival of sepsis by the down-regulation of inflammation-related genes (such e.g. IL-10 and IL-6) and a shift towards the up-regulation of genes involved in promoting phagocytosis and bacterial killing 104. The direct interaction of MSCs with the oral bacteria F. nucleatum and P. gingivalis led to a lower secretion of IL-8 compared to a differentiated tumour cell line 66. Raffaghello et al. support the immunomodulatory function of MSCs showing the inhibition of neutrophil apoptosis because of the secretion of IL-6 by MSCs 105. Nevertheless, these results should be interpreted carefully as it is speculated that the cytokine induction profile of stem cells is dependent on the cell type, bacterial species and methodology used (e.g. period of stimulation) 79,106.

Current challenges and future perspectives

The rapid advancements in the field of dental research over the last few years could realize the promise of tissue regeneration through stem cells. Specifically, the demand for novel therapies against inflammatory diseases, like periodontitis, has created the need for a better understanding of the behaviour of the multipotent cells at sites of infection. Stem cells are supposed to support tissue homoeostasis by providing soluble factors, transdifferentiation or cell fusion 107. Hence, studies demonstrating stem cell responsiveness to bacteria raise questions on the possible contribution of multipotent cells to both tissue regeneration and outbreak of inflammation. Selected reports on the impact of bacteria on stem cells are listed in Table1.

Table 1.

Selected reports on the impact of bacteria on stem cells

Biological impact Cell populations Bacterial species Experimental model Reference
Cell viability DFPCs P. gingivalis Treatment with LPS 47
PDLSCs P. gingivalis Treatment with LPS 53
DFPCs, BMSCs P. gingivalis Treatment with LPS 78
USSCs Undefined Treatment with LPS and flagellin 79
DPSCs S. mutans Treatment with LPS 80
Differentiation DFPCs P. gingivalis Treatment with LPS 47
BMSCs E. coli Treatment with LPS 52
PDLSCs P. gingivalis Treatment with LPS 53
USSCs Undefined Treatment with LPS and flagellin 79
DPPCs A.actinomyce-temcomitans Treatment with LPS 87
DPSCs P. gingivalis Treatment with LPS 88
Immunomodulation DFPCs P. gingivalis Treatment with LPS 47
BMSCs P. gingivalis, F. nucleatum, A.actinomyce-temcomitans Co-culture model 69
DFPCs, BMSCs P. gingivalis, F. nucleatum Co-culture model 70
DFPCs, BMSCs P. gingivalis Treatment with LPS 78
USSCs Undefined Treatment with LPS and flagellin 79
ESC-ECs Undefined Treatment with LPS and C12-iE-DAP 99
DFSCs, DPSCs Undefined Treatment with LPS 100
AMSCs S. typhimurium, L. acidophilus Co-culture model 101
DPSCs P. gingivalis, E. coli, P. endodontalis Treatment with LPS 102
DPSCs Undefined Treatment with LPS 103
MSCs Undefined Polymicrobial model of sepsis 104

Stem cells: AMSCs, adipose-derived mesenchymal stem cells; BMSCs, bone marrow stem cells; DFPCs, dental follicle progenitors cells; DPPCs, dental pulp progenitor cells; DPSCs, dental pulp stem cells; ESC-ECs, human embryonic stem cell-derived endothelial cells; MSCs, mesenchymal stem cells; PDLSCs, periodontal ligament stem cells; USSCs, unrestricted somatic stem cells. Bacteria: A. actinomycetemcomitans, Aggregatibacter actinomycetemcomitans; E. coli, Escherichia coli; F. Prausnitzii, Faecalibacterium prausnitzii; L. acidophilus, Lactobacillus acidophilus; P. endodontalis, Porphyromonas endodontalis; P. gingivalis, Porphyromonas gingivalis; S. mutans, Streptococcus mutans; S. typhimurium, Salmonella typhimurium.

The notion that bacteria may stimulate and drive the regenerative potential of stem cells should be further explored. Till now, data from in vitro studies utilizing single populations of cells challenged with bacterial components or mono-infections of planktonic bacteria may not adequately portray human periodontal diseases. Another significant parameter, which should be taken into consideration, is the oxygen concentration of in vitro models, as most of the pathogenic species implicated in the pathogenesis of periodontitis are obligate anaerobes. It is also remarkable that only few studies have used PDLSCs, which are the main population of multipotent cells residing within the periodontium. Thus, the development of new experimental settings to better resemble the in vivo periodontal milieu seems to be crucial.

A better understanding of the beneficial effects of bacteria on stem cells may allow future interventions based on cell priming with bacterial components prior to transplantation in sites of tissue destruction. Even the colonization of inflamed tissues with specific bacterial species that promote the mobilization of tissue-resident multipotent cell populations could be part of new therapeutic approaches. On the other side, the extent of stem cells’ involvement in immunomodulation remains to be clarified. Both immunosuppression and stimulation of host immune responses regulated by stem cells could be used as advanced tools against bacterially induced inflammation. In conclusion, the identification of intracellular signalling pathways regulating multipotency and immunomodulation of stem cells being exposed to bacteria may enable the development of successful therapeutic interventions in inflammatory diseases.

Conflicts of interest

The authors confirm that there are no conflicts of interest.

Author contributions

Kyriaki Chatzivasileiou and Katja Kriebel contributed substantially to the conception and design of the study and wrote the paper. Hermann Lang, Bernd Kreikemeyer and Gustav Steinhoff contributed to the conception and critical revision of the article and provided the final approval of the version to be published.

References

  1. Nemeth K, Mayer B, Mezey E. Modulation of bone marrow stromal cell functions in infectious diseases by toll-like receptor ligands. J Mol Med. 2010;88:5–10. doi: 10.1007/s00109-009-0523-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Kolb-Mäurer A, Goebel W. Susceptibility of hematopoietic stem cells to pathogens: role in virus/bacteria tropism and pathogenesis. FEMS Microbiol Lett. 2003;226:203–7. doi: 10.1016/S0378-1097(03)00643-8. [DOI] [PubMed] [Google Scholar]
  3. Auletta JJ, Deans RJ, Bartholomew AM. Emerging roles for multipotent, bone marrow-derived stromal cells in host defense. Blood. 2012;119:1801–9. doi: 10.1182/blood-2011-10-384354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Zarco MF, Vess TJ, Ginsburg GS. The oral microbiome in health and disease and the potential impact on personalized dental medicine. Oral Dis. 2012;18:109–20. doi: 10.1111/j.1601-0825.2011.01851.x. [DOI] [PubMed] [Google Scholar]
  5. Paster BJ, Olsen I, Aas JA, et al. The breadth of bacterial diversity in the human periodontal pocket and other oral sites. Periodontol 2000. 2006;42:80–7. doi: 10.1111/j.1600-0757.2006.00174.x. [DOI] [PubMed] [Google Scholar]
  6. Listgarten MA. Structure of the microbial flora associated with periodontal health and disease in man. A light and electron microscopic study. J Periodontol. 1976;47:1–18. doi: 10.1902/jop.1976.47.1.1. [DOI] [PubMed] [Google Scholar]
  7. Avila M, Ojcius DM, Yilmaz O. The oral microbiota: living with a permanent guest. DNA Cell Biol. 2009;28:405–11. doi: 10.1089/dna.2009.0874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Jain S, Darveau RP. Contribution of Porphyromonas gingivalis lipopolysaccharide to periodontitis. Periodontol 2000. 2010;54:53–70. doi: 10.1111/j.1600-0757.2009.00333.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Socransky SS, Haffajee AD. Periodontal microbial ecology. Periodontol 2000. 2005;38:135–87. doi: 10.1111/j.1600-0757.2005.00107.x. [DOI] [PubMed] [Google Scholar]
  10. Dewhirst FE, Chen T, Izard J, et al. The human oral microbiome. J Bacteriol. 2010;192:5002–17. doi: 10.1128/JB.00542-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Aas JA, Paster BJ, Stokes LN, et al. Defining the normal bacterial flora of the oral cavity. J Clin Microbiol. 2005;43:5721–32. doi: 10.1128/JCM.43.11.5721-5732.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Zaura E, Keijser BJ, Huse SM, et al. Defining the healthy “core microbiome” of oral microbial communities. BMC Microbiol. 2009;9:259. doi: 10.1186/1471-2180-9-259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bik EM, Long CD, Armitage GC, et al. Bacterial diversity in the oral cavity of 10 healthy individuals. ISME J. 2010;4:962–74. doi: 10.1038/ismej.2010.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Marsh PD, Percival RS. The oral microflora–friend or foe? Can we decide? Int Dent J. 2006;56:233–9. doi: 10.1111/j.1875-595x.2006.tb00107.x. [DOI] [PubMed] [Google Scholar]
  15. Filoche S, Wong L, Sissons CH. Oral biofilms: emerging concepts in microbial ecology. J Dent Res. 2010;89:8–18. doi: 10.1177/0022034509351812. [DOI] [PubMed] [Google Scholar]
  16. Ruby EG. Symbiotic conversations are revealed under genetic interrogation. Nat Rev Microbiol. 2008;6:752–62. doi: 10.1038/nrmicro1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Parahitiyawa NB, Scully C, Leung WK, et al. Exploring the oral bacterial flora: current status and future directions. Oral Dis. 2010;16:136–45. doi: 10.1111/j.1601-0825.2009.01607.x. [DOI] [PubMed] [Google Scholar]
  18. Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal diseases. Lancet. 2005;366:1809–20. doi: 10.1016/S0140-6736(05)67728-8. [DOI] [PubMed] [Google Scholar]
  19. Van Dyke TE. The management of inflammation in periodontal disease. J Periodontol. 2008;79:1601–8. doi: 10.1902/jop.2008.080173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Dewhirst FE, Tamer MA, Ericson RE, et al. The diversity of periodontal spirochetes by 16S rRNA analysis. Oral Microbiol Immunol. 2000;15:196–202. doi: 10.1034/j.1399-302x.2000.150308.x. [DOI] [PubMed] [Google Scholar]
  21. Socransky SS, Haffajee AD. Evidence of bacterial etiology: a historical perspective. Periodontol 2000. 1994;5:7–25. doi: 10.1111/j.1600-0757.1994.tb00016.x. [DOI] [PubMed] [Google Scholar]
  22. Page RC. The role of inflammatory mediators in the pathogenesis of periodontal disease. J Periodontal Res. 1991;26:230–42. doi: 10.1111/j.1600-0765.1991.tb01649.x. [DOI] [PubMed] [Google Scholar]
  23. Siqueira JF, Jr, Rôças IN. Bacterial pathogenesis and mediators in apical periodontitis. Braz Dent J. 2007;18:267–80. doi: 10.1590/s0103-64402007000400001. [DOI] [PubMed] [Google Scholar]
  24. Noreen M, Shah MA, Mall SM, et al. TLR4 polymorphisms and disease susceptibility. Inflamm Res. 2012;61:177–88. doi: 10.1007/s00011-011-0427-1. [DOI] [PubMed] [Google Scholar]
  25. Lindhe J, Westfelt E, Nyman S, et al. Long-term effect of surgical/non-surgical treatment of periodontal disease. J Clin Periodontol. 1984;11:448–58. doi: 10.1111/j.1600-051x.1984.tb01344.x. [DOI] [PubMed] [Google Scholar]
  26. Badersten A, Nilveus R, Egelberg J. Effect of nonsurgical periodontal therapy (VIII). Probing attachment changes related to clinical characteristics. J Clin Periodontol. 1987;14:425–32. doi: 10.1111/j.1600-051x.1987.tb01548.x. [DOI] [PubMed] [Google Scholar]
  27. Kaldahl WB, Kalkwarf KL, Patil KD, et al. Evaluation of four modalities of periodontal therapy. Mean probing depth, probing attachment level and recession changes. J Periodontol. 1988;59:783–93. doi: 10.1902/jop.1988.59.12.783. [DOI] [PubMed] [Google Scholar]
  28. Caton JG. Overview of clinical trials on periodontal regeneration. Ann Periodontol. 1997;2:215–22. doi: 10.1902/annals.1997.2.1.215. [DOI] [PubMed] [Google Scholar]
  29. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  30. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3:393–403. doi: 10.1111/j.1365-2184.1970.tb00347.x. [DOI] [PubMed] [Google Scholar]
  31. Friedenstein AJ, Chailakhyan RK, Latsinik NV, et al. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation. 1974;17:331–40. doi: 10.1097/00007890-197404000-00001. [DOI] [PubMed] [Google Scholar]
  32. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95. doi: 10.1091/mbc.E02-02-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Roubelakis MG, Bitsika V, Zagoura D, et al. In vitro and in vivo properties of distinct populations of amniotic fluid mesenchymal progenitor cells. J Cell Mol Med. 2011;15:1896–913. doi: 10.1111/j.1582-4934.2010.01180.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Fiegel HC, Lange C, Kneser U, et al. Fetal and adult liver stem cells for liver regeneration and tissue engineering. J Cell Mol Med. 2006;10:577–87. doi: 10.1111/j.1582-4934.2006.tb00422.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Kögler G, Sensken S, Airey JA, et al. A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. J Exp Med. 2004;200:123–35. doi: 10.1084/jem.20040440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Gronthos S, Mankani M, Brahim J, et al. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA. 2000;97:13625–30. doi: 10.1073/pnas.240309797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Morsczeck C, Götz W, Schierholz J, et al. Isolation of precursor cells (PCs) from human dental follicle of wisdom teeth. Matrix Biol. 2005;24:155–65. doi: 10.1016/j.matbio.2004.12.004. [DOI] [PubMed] [Google Scholar]
  38. Miura M, Gronthos S, Zhao M, et al. SHED: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci USA. 2003;100:5807–12. doi: 10.1073/pnas.0937635100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Sonoyama W, Liu Y, Fang D, et al. Mesenchymal stem cell-mediated functional tooth regeneration in swine. PLoS ONE. 2006;1:e79. doi: 10.1371/journal.pone.0000079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Chen FM, Sun HH, Lu H, et al. Stem cell-delivery therapeutics for periodontal tissue regeneration. Biomaterials. 2012;33:6320–44. doi: 10.1016/j.biomaterials.2012.05.048. [DOI] [PubMed] [Google Scholar]
  41. Lindroos B, Mäenpää K, Ylikomi T, et al. Characterisation of human dental stem cells and buccal mucosa fibroblasts. Biochem Biophys Res Commun. 2008;368:329–35. doi: 10.1016/j.bbrc.2008.01.081. [DOI] [PubMed] [Google Scholar]
  42. Huang GT, Gronthos S, Shi S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. J Dent Res. 2009;88:792–806. doi: 10.1177/0022034509340867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Kim BC, Bae H, Kwon IK, et al. Osteoblastic/cementoblastic and neural differentiation of dental stem cells and their applications to tissue engineering and regenerative medicine. Tissue Eng Part B Rev. 2012;18:235–44. doi: 10.1089/ten.teb.2011.0642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Bartold PM, Shi S, Gronthos S. Stem cells and periodontal regeneration. Periodontol 2000. 2006;40:164–72. doi: 10.1111/j.1600-0757.2005.00139.x. [DOI] [PubMed] [Google Scholar]
  45. Ivanovski S, Gronthos S, Shi S, et al. Stem cells in the periodontal ligament. Oral Dis. 2006;12:358–63. doi: 10.1111/j.1601-0825.2006.01253.x. [DOI] [PubMed] [Google Scholar]
  46. Trofin EA, Monsarrat P, Kémoun P. Cell therapy of periodontium: from animal to human? Front Physiol. 2013;4:325. doi: 10.3389/fphys.2013.00325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Morsczeck CO, Drees J, Gosau M. Lipopolysaccharide from Escherichia coli but not from Porphyromonas gingivalis induce pro-inflammatory cytokines and alkaline phosphatase in dental follicle cells. Arch Oral Biol. 2012;57:1595–601. doi: 10.1016/j.archoralbio.2012.07.016. [DOI] [PubMed] [Google Scholar]
  48. Sorrell JM, Caplan AI. Topical delivery of mesenchymal stem cells and their function in wounds. Stem Cell Res Ther. 2010;1:30. doi: 10.1186/scrt30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol. 2007;213:341–7. doi: 10.1002/jcp.21200. [DOI] [PubMed] [Google Scholar]
  50. Kinane DF, Lappin DF. Immune processes in periodontal disease: a review. Ann Periodontol. 2002;7:62–71. doi: 10.1902/annals.2002.7.1.62. [DOI] [PubMed] [Google Scholar]
  51. Kebschull M, Papapanou PN. Periodontal microbial complexes associated with specific cell and tissue responses. J Clin Periodontol. 2011;38:17–27. doi: 10.1111/j.1600-051X.2010.01668.x. [DOI] [PubMed] [Google Scholar]
  52. Mo IF, Yip KH, Chan WK, et al. Prolonged exposure to bacterial toxins downregulated expression of toll-like receptors in mesenchymal stromal cell-derived osteoprogenitors. BMC Cell Biol. 2008;9:52. doi: 10.1186/1471-2121-9-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Kato H, Taguchi Y, Tominaga K, et al. Porphyromonas gingivalis LPS inhibits osteoblastic differentiation and promotes pro-inflammatory cytokine production in human periodontal ligament stem cells. Arch Oral Biol. 2014;59:167–75. doi: 10.1016/j.archoralbio.2013.11.008. [DOI] [PubMed] [Google Scholar]
  54. Stathopoulou PG, Galicia JC, Benakanakere MR, et al. Porphyromonas gingivalis induce apoptosis in human gingival epithelial cells through a gingipain-dependent mechanism. BMC Microbiol. 2009;9:107. doi: 10.1186/1471-2180-9-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Laheij AM, de Soet JJ, Veerman EC, et al. The influence of oral bacteria on epithelial cell migration in vitro. Mediators Inflamm. 2013;2013:154532. doi: 10.1155/2013/154532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Haffajee AD, Socransky SS. Microbial etiological agents of destructive periodontal diseases. Periodontol 2000. 1994;5:78–111. doi: 10.1111/j.1600-0757.1994.tb00020.x. [DOI] [PubMed] [Google Scholar]
  57. Bostanci N, Allaker RP, Belibasakis GN, et al. Porphyromonas gingivalis antagonises Campylobacter rectus induced cytokine production by human monocytes. Cytokine. 2007;39:147–56. doi: 10.1016/j.cyto.2007.07.002. [DOI] [PubMed] [Google Scholar]
  58. Yin L, Swanson B, An J, et al. Differential effects of periopathogens on host protease inhibitors SLPI, elafin, SCCA1, and SCCA2. J Oral Microbiol. 2010;2 doi: 10.3402/jom.v2i0.5070. doi: 10.3402/jom.v2i0.5070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Adams CA. The probiotic paradox: live and dead cells are biological response modifiers. Nutr Res Rev. 2010;23:37–46. doi: 10.1017/S0954422410000090. [DOI] [PubMed] [Google Scholar]
  60. Guggenheim B, Gmür R, Galicia JC, et al. In vitro modeling of host-parasite interactions: the ‘subgingival’ biofilm challenge of primary human epithelial cells. BMC Microbiol. 2009;9:280. doi: 10.1186/1471-2180-9-280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Zijnge V, van Leeuwen MB, Degener JE, et al. Oral biofilm architecture on natural teeth. PLoS ONE. 2010;5:e9321. doi: 10.1371/journal.pone.0009321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Sissons CH. Artificial dental plaque biofilm model systems. Adv Dent Res. 1997;11:110–26. doi: 10.1177/08959374970110010201. [DOI] [PubMed] [Google Scholar]
  63. Park JH, Lee JK, Um HS, et al. A periodontitis-associated multispecies model of an oral biofilm. J Periodontal Implant Sci. 2014;44:79–84. doi: 10.5051/jpis.2014.44.2.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Tabenski L, Maisch T, Santarelli F, et al. Individual growth detection of bacterial species in an in vitro oral polymicrobial biofilm model. Arch Microbiol. 2014;196:819–28. doi: 10.1007/s00203-014-1021-z. [DOI] [PubMed] [Google Scholar]
  65. Li L, Michel R, Cohen J, et al. Intracellular survival and vascular cell-to-cell transmission of Porphyromonas gingivalis. BMC Microbiol. 2008;8:26. doi: 10.1186/1471-2180-8-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Saito A, Inagaki S, Ishihara K. Differential ability of periodontopathic bacteria to modulate invasion of human gingival epithelial cells by Porphyromonas gingivalis. Microb Pathog. 2009;47:329–33. doi: 10.1016/j.micpath.2009.09.012. [DOI] [PubMed] [Google Scholar]
  67. Dabija-Wolter G, Sapkota D, Cimpan MR, et al. Limited in-depth invasion of Fusobacterium nucleatum into in vitro reconstructed human gingiva. Arch Oral Biol. 2012;57:344–51. doi: 10.1016/j.archoralbio.2011.09.015. [DOI] [PubMed] [Google Scholar]
  68. Maier E, Anderson RC, Roy NC. Understanding how commensal obligate anaerobic bacteria regulate immune functions in the large intestine. Nutrients. 2014;7:45–73. doi: 10.3390/nu7010045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kriebel K, Biedermann A, Kreikemeyer B, et al. Anaerobic co-culture of mesenchymal stem cells and anaerobic pathogens - a new in vitro model system. PLoS ONE. 2013;8:e78226. doi: 10.1371/journal.pone.0078226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Biedermann A, Kriebel K, Kreikemeyer B, et al. Interactions of anaerobic bacteria with dental stem cells: an in vitro study. PLoS ONE. 2014;9:e110616. doi: 10.1371/journal.pone.0110616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Ulluwishewa D, Anderson RC, Young W, et al. Live Faecalibacterium prausnitzii in an apical anaerobic model of the intestinal epithelial barrier. Cell Microbiol. 2015;17:226–40. doi: 10.1111/cmi.12360. [DOI] [PubMed] [Google Scholar]
  72. Yamasaki M, Nakata K, Imaizumi I, et al. Cytotoxic effect of endodontic bacteria on periapical fibroblasts. J Endod. 1998;24:534–9. doi: 10.1016/S0099-2399(98)80072-1. [DOI] [PubMed] [Google Scholar]
  73. Tabeta K, Yamazaki K, Akashi S, et al. Toll-like receptors confer responsiveness to lipopolysaccharide from Porphyromonas gingivalis in human gingival fibroblasts. Infect Immun. 2000;68:3731–5. doi: 10.1128/iai.68.6.3731-3735.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Wang PL, Oido-Mori M, Fujii T, et al. Heterogeneous expression of Toll-like receptor 4 and downregulation of Toll-like receptor 4 expression on human gingival fibroblasts by Porphyromonas gingivalis lipopolysaccharide. Biochem Biophys Res Commun. 2001;288:863–7. doi: 10.1006/bbrc.2001.5842. [DOI] [PubMed] [Google Scholar]
  75. Pevsner-Fischer M, Morad V, Cohen-Sfady M, et al. Toll-like receptors and their ligands control mesenchymal stem cell functions. Blood. 2007;109:1422–32. doi: 10.1182/blood-2006-06-028704. [DOI] [PubMed] [Google Scholar]
  76. Jiang H, Patel PH, Kohlmaier A, et al. Cytokine/Jak/Stat signaling mediates regeneration and homeostasis in the Drosophila midgut. Cell. 2009;137:1343–55. doi: 10.1016/j.cell.2009.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Buchon N, Broderick NA, Poidevin M, et al. Drosophila intestinal response to bacterial infection: activation of host defense and stem cell proliferation. Cell Host Microbe. 2009;5:200–11. doi: 10.1016/j.chom.2009.01.003. [DOI] [PubMed] [Google Scholar]
  78. Chatzivasileiou K, Lux CA, Steinhoff G, et al. Dental follicle progenitor cells responses to Porphyromonas gingivalis LPS. J Cell Mol Med. 2013;17:766–73. doi: 10.1111/jcmm.12058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. van den Berk LC, Jansen BJH, Siebers-Vermeulen KG, et al. Toll-like receptor triggering in cord blood mesenchymal stem cells. J Cell Mol Med. 2009;13:3415–26. doi: 10.1111/j.1582-4934.2009.00653.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Liu Y, Gao Y, Zhan X, et al. TLR4 activation by lipopolysaccharide and Streptococcus mutans induces differential regulation of proliferation and migration in human dental pulp stem cells. J Endod. 2014;40:1375–81. doi: 10.1016/j.joen.2014.03.015. [DOI] [PubMed] [Google Scholar]
  81. Yilmaz O. The chronicles of Porphyromonas gingivalis: the microbium, the human oral epithelium and their interplay. Microbiology. 2008;154:2897–903. doi: 10.1099/mic.0.2008/021220-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Hynes K, Menicanin D, Gronthos S, et al. Clinical utility of stem cells for periodontal regeneration. Periodontol 2000. 2012;59:203–27. doi: 10.1111/j.1600-0757.2012.00443.x. [DOI] [PubMed] [Google Scholar]
  83. Xiao L, Tsutsui T. Human dental mesenchymal stem cells and neural regeneration. Hum Cell. 2013;26:91–6. doi: 10.1007/s13577-013-0069-4. [DOI] [PubMed] [Google Scholar]
  84. Ronay V, Belibasakis GN, Attin T, et al. Expression of embryonic stem cell markers and osteogenic differentiation potential in cells derived from periodontal granulation tissue. Cell Biol Int. 2014;38:179–86. doi: 10.1002/cbin.10190. [DOI] [PubMed] [Google Scholar]
  85. Abe S, Imaizumi M, Mikami Y, et al. Oral bacterial extracts facilitate early osteogenic/dentinogenic differentiation in human dental pulp-derived cells. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2010;109:149–54. doi: 10.1016/j.tripleo.2009.08.028. [DOI] [PubMed] [Google Scholar]
  86. Morozumi A. High concentration of sodium butyrate suppresses osteoblastic differentiation and mineralized nodule formation in ROS17/2.8 cells. J Oral Sci. 2011;53:509–16. doi: 10.2334/josnusd.53.509. [DOI] [PubMed] [Google Scholar]
  87. Nomiyama K, Kitamura C, Tsujisawa T, et al. Effects of lipopolysaccharide on newly established rat dental pulp-derived cell line with odontoblastic properties. J Endod. 2007;33:1187–91. doi: 10.1016/j.joen.2007.05.015. [DOI] [PubMed] [Google Scholar]
  88. Yamagishi VT, Torneck CD, Friedman S, et al. Blockade of TLR2 inhibits Porphyromonas gingivalis suppression of mineralized matrix formation by human dental pulp stem cells. J Endod. 2011;37:812–8. doi: 10.1016/j.joen.2011.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Hirose K, Isogai E, Mizugai H, et al. Inductive effect of Porphyromonas gingivalis fimbriae on differentiation of human monocytic tumor cell line U937. Oral Microbiol Immunol. 1996;11:62–4. doi: 10.1111/j.1399-302x.1996.tb00338.x. [DOI] [PubMed] [Google Scholar]
  90. Zhang W, Swearingen EB, Ju J, et al. Porphyromonas gingivalis invades osteoblasts and inhibits bone formation. Microbes Infect. 2010;12:838–45. doi: 10.1016/j.micinf.2010.05.011. [DOI] [PubMed] [Google Scholar]
  91. Li Z, Jiang CM, An S, et al. Immunomodulatory properties of dental tissue-derived mesenchymal stem cells. Oral Dis. 2014;20:25–34. doi: 10.1111/odi.12086. [DOI] [PubMed] [Google Scholar]
  92. Meisel R, Zibert A, Laryea M, et al. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004;103:4619–21. doi: 10.1182/blood-2003-11-3909. [DOI] [PubMed] [Google Scholar]
  93. Sotiropoulou PA, Perez SA, Gritzapis AD, et al. Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells. 2006;24:74–85. doi: 10.1634/stemcells.2004-0359. [DOI] [PubMed] [Google Scholar]
  94. Sato K, Ozaki K, Oh I, et al. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 2007;109:228–34. doi: 10.1182/blood-2006-02-002246. [DOI] [PubMed] [Google Scholar]
  95. Selmani Z, Naji A, Zidi I, et al. 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. 2008;26:212–22. doi: 10.1634/stemcells.2007-0554. [DOI] [PubMed] [Google Scholar]
  96. English K, Ryan JM, Tobin L, et al. 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. 2009;156:149–60. doi: 10.1111/j.1365-2249.2009.03874.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Ren G, Su J, Zhang L, et al. Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells. 2009;27:1954–62. doi: 10.1002/stem.118. [DOI] [PubMed] [Google Scholar]
  98. Zhang Q, Shi S, Liu Y, et al. Mesenchymal stem cells derived from human gingiva are capable of immunomodulatory functions and ameliorate inflammation-related tissue destruction in experimental colitis. J Immunol. 2009;183:7787–98. doi: 10.4049/jimmunol.0902318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Reed DM, Foldes G, Gatheral T, et al. Pathogen sensing pathways in human embryonic stem cell derived-endothelial cells: role of NOD1 receptors. PLoS ONE. 2014;9:e91119. doi: 10.1371/journal.pone.0091119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Tomic S, Djokic J, Vasilijic S, et al. Immunomodulatory properties of mesenchymal stem cells derived from dental pulp and dental follicle are susceptible to activation by toll-like receptor agonists. Stem Cells Dev. 2011;20:695–708. doi: 10.1089/scd.2010.0145. [DOI] [PubMed] [Google Scholar]
  101. Kol A, Foutouhi S, Walker NJ, et al. Gastrointestinal microbes interact with canine adipose-derived mesenchymal stem cells in vitro and enhance immunomodulatory functions. Stem Cells Dev. 2014;23:1831–43. doi: 10.1089/scd.2014.0128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Chang J, Zhang C, Tani-Ishii N, et al. NF-kappaB activation in human dental pulp stem cells by TNF and LPS. J Dent Res. 2005;84:994–8. doi: 10.1177/154405910508401105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. He W, Qu T, Yu Q, et al. LPS induces IL-8 expression through TLR4, MyD88, NF-kappaB and MAPK pathways in human dental pulp stem cells. Int Endod J. 2013;46:128–36. doi: 10.1111/j.1365-2591.2012.02096.x. [DOI] [PubMed] [Google Scholar]
  104. Mei SH, Haitsma JJ, Dos Santos CC, et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med. 2010;182:1047–57. doi: 10.1164/rccm.201001-0010OC. [DOI] [PubMed] [Google Scholar]
  105. Raffaghello L, Bianchi G, Bertolotto M, et al. Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells. 2008;26:151–62. doi: 10.1634/stemcells.2007-0416. [DOI] [PubMed] [Google Scholar]
  106. Feng Z, Weinberg A. Role of bacteria in health and disease of periodontal tissues. Periodontol 2000. 2006;40:50–76. doi: 10.1111/j.1600-0757.2005.00148.x. [DOI] [PubMed] [Google Scholar]
  107. Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal stromal cells. Blood. 2007;110:3499–506. doi: 10.1182/blood-2007-02-069716. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

RESOURCES