Skip to main content

Genetic contribution to mesenchymal stem cell dysfunction in systemic lupus erythematosus

Abstract

Allogeneic mesenchymal stem cell (MSC) transplantation has recently become a promising therapy for patients with systemic lupus erythematosus (SLE). MSCs are a kind of multipotent stem cell than can efficiently modulate both innate and adaptive immune responses, yet those from SLE patients themselves fail to maintain the balance of immune cells, which is partly due to the abnormal genetic background. Clarifying genetic factors associated with MSC dysfunction may be helpful to delineate SLE pathogenesis and provide new therapeutic targets. In this review, the scientific evidence on the genetic contribution to MSC dysfunction in SLE is summarized.

Background

Mesenchymal stem cells (MSCs), mainly located in the adult bone marrow, have a capacity for self-renewal and potential for multilineage differentiation into a variety of cell types, such as osteoblasts, chondrocytes, myocytes, and adipocytes [1]. These cells also provide a supportive microenvironment for hematopoietic stem cells, to maintain their growth, differentiation, and proper function [2]. Recently, MSCs have been demonstrated to display profound immunosuppressive effects on various immune cells [3] and thus are considered as a new therapeutic paradigm for autoimmune diseases.

Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by the activation of both innate and adaptive immune responses. Although survival of SLE patients has improved considerably in the past decades, a substantial proportion of patients are still refractory to routine treatments and expected to have a poor prognosis. While allogeneic MSC transplantation results in the induction of clinical remission and improvement in organ dysfunction for patients with refractory SLE [4], evidence has shown that autologous MSCs are not beneficial for these patients [5], implying that abnormalities of MSCs themselves is involved in the progress of SLE disease.

As bone marrow (BM) MSCs separated from the in vivo environment of SLE still present impaired capacity to inhibit immune cells and induce peripheral tolerance [6], it is reasonable to mainly attribute MSC dysfunction to the intrinsic genetic defects in this disease. To further understand the regulatory mechanism of SLE MSCs, possible genetic factors implicated in the regulation of MSC function are discussed in combination with the latest research findings.

Immunomodulatory effect of MSCs

Animal model studies and phase I/II clinical trials have demonstrated that MSCs have therapeutic potential in the treatment of a variety of autoimmune diseases, including SLE [7,8,9,10]. Although MSCs from different sources exhibited various immunomodulatory capacities in vitro [11], their efficacy is similar as reported in a clinical study using BM or umbilical cord-derived MSC transplantation for severe and refractory SLE patients [4]. However, the mechanisms by which MSCs affect immune cells and the underlying signaling pathways remain unclear. Recently, it has been proposed that MSCs may act through both paracrine secretion of soluble factors and cell–cell contact.

MSCs are associated with the inhibition of T-cell proliferation and upregulation of T regulatory (Treg) cells [12]. Evidence shows that murine MSCs secrete nitric oxide (NO) to inhibit T-cell production [13], and MSCs from mice with inducible nitric oxide synthase (iNOS) knocked down display impaired ability to prevent T-cell proliferation [14]. Besides, iNOS activation in MSCs has been shown to greatly inhibit the expansion of T follicular helper (Tfh) cells, a CD4+ T-cell subtype that helps B cells to generate affinity-matured antibodies, in lupus mice [15]. In human MSCs, the effects of NO is replaced by indoleamine 2,3-dioxygenase (IDO) [16], the production of which is enhanced by CD8+ T-cell-secreted interferon-γ (IFN-γ) [17], suggesting that different species may employ different effector molecules. Different from other T-cell subsets, CD4+CD25+FoxP3+ Treg cell levels are elevated after MSCs transplantation [5]. These cells could be induced from purified CD4+ T cells by allogeneic human MSCs in vitro, as mediated by prostaglandin E2 (PGE2) and transforming growth factor beta1 (TGF-β1) [18]. Through the upregulation of interleukin-10 (IL-10) and Fas ligand, CD8+ Treg cells could also be proliferated and functionally enhanced by MSCs [19]. Besides these soluble factors, cell–cell contact also plays an important role in MSC-induced T-cell regulation [15]. Recently, MSCs have been shown to be capable of transferring mitochondria into T cells [20], which may provide an explanation for how cell–cell contact regulates the immunomodulatory functions of MSCs.

MSCs participate in inhibiting B cell proliferation, differentiation, antibody production, and apoptosis [21]. It has been proven that MSCs stimulated with IFN-γ and tumor necrosis factor α (TNF-α) have enhanced regulatory effects on B cells [22]. Through programmed death 1(PD-1) and programmed death ligand-1 (PDL-1) interaction, IFN-γ may increase the amount of PDL-1 in MSCs to inhibit the proliferation and differentiation of B cells [23]. Meanwhile, MSC-derived chemokine C-C motif ligand 2 (CCL2) is also involved in B-cell proliferation, plasma cell differentiation, and immunoglobulin generation through the inactivation of signal transducer and activator of transcription 3 (STAT3) and induction of paired box protein 5 [24]. CCL2 was originally considered as a pro-inflammatory chemokine that helps recruit monocytes or macrophages to areas of inflammation. However, MSCs could turn this chemokine into an anti-inflammatory phenotype though matrix metalloproteinase (MMP)-induced proteolysis.

MSCs can reprogram macrophages and enhance their anti-inflammatory function [25, 26]. Macrophages are capable of being educated into alternatively activated macrophages (AAMs; also called M2), which possess enhanced phagocytic activity when co-cultured with MSCs, via up-regulation of IL-10/TGF-β1 and down-regulation of TNF-α/iNOS [27, 28]. These M2 macrophages express high levels of CD206 and have potent immunoregulatory function, characterized by enhanced inhibition of CD4+ T-cell differentiation and decreased ability to induce Th17 cell expansion. Recently, a role for IL-6 in MSC-driven macrophage polarization has been described in SLE patients [29].

Other immune cells, such as dendritic cells (DCs) and natural killer (NK) cells, are also regulated by MSCs. The maturation and antigen-presenting functions of DCs are suppressed by MSCs via soluble factors such as PGE2 and IL6 [30,31,32]. Meanwhile, IDO and PGE2 secreted by MSCs play a key role in MSC-induced inhibition of NK cells [33]. Interestingly, none of these soluble factors is solely responsible for the entire immune regulatory network of MSCs. Instead, a combination of molecules is usually required for MSC-mediated treatment effects.

Impaired immune regulation of SLE MSCs

BM MSCs derived from SLE patients exhibit a lot of abnormalities compared with normal controls, including cytoskeleton-related defects [34] and increased cell senescence as well as apoptosis [35]. Their potential for differentiation and migration is greatly impaired [36]. Moreover, expression profiles of genes related to immune function in SLE MSCs, such as IDO, IL-6, IL-7, and TGF-β, are distinct from those in normal subjects [17, 37,38,39]. Correspondingly, the function of MSCs from SLE patients or lupus animal models is severely damaged and not sufficient to suppress various immune cells [40, 41], and may thus contribute to the onset of autoimmunity.

In SLE, due to the lack of efficient MSC regulation, the balance between Treg and Tfh cells is usually broken. As a consequence, peripheral Treg cells are decreased and Tfh cells are increased, especially in those with active disease [15, 38]. Evidence has shown that allogeneic MSCs could increase the number of peripheral Treg cells through the secretion of TGF-β in SLE patients [5, 42] and inhibit Tfh cell expansion via the activation of iNOS in lupus mice [15], indicating there is a restoration of Treg/Tfh balance after MSC treatment.

Similar to T cells, BM MSCs from SLE patients and lupus mice are less effective at preventing the proliferation and inhibition of B cells, as well as the secretion of autoantibodies, which is mainly mediated by reduced CCL2 production [6]. After the knock-down of the CCL2 gene, the ability of MSCs to suppress B cells is greatly impaired. Meanwhile, over-expression of CCL2 enhances the efficacy of MSCs to regulate B-cell proliferation, along with ameliorated renal pathology and reduced autoantibody production [6]. Besides CCL2, there is evidence showing that mice lacking PD-1 are capable of developing an SLE-like disease [43]. PD-1 reduction may hinder the cell-to-cell contact between MSCs and B cells and consequently inhibit B cell proliferation and differentiation [23].

Besides acquired immunity, the abnormality of innate immunity in SLE is also related to MSCs [40]. Macrophages from either lupus mice or SLE patients display decreased CD206 expression and inefficient phagocytic activity that could be corrected by MSCs from a healthy population in an IL-6-dependent manner [29]. Since IL-6 expression is reduced in BM MSCs from SLE patients [39], it is not surprising to find that SLE MSCs fail to regulate macrophage over-activation in vivo. Accordingly, impaired phagocytic activity of macrophages may result in the accumulation of apoptotic debris and lead to sequelae of autoimmune phenomena [44].

Recently the effect of MSCs on autophagy has been revealed. T cells from SLE patients appear to have higher levels of autophagy [20, 45], a basic cellular homeostatic process that enables cells to eliminate portions of their own cytoplasmic contents. Abnormal autophagic activity in SLE T cells is caused primarily by cellular accumulation of functionally defective mitochondria, which activates the autophagy process and subsequently promotes apoptosis of T cells [46]. Meanwhile, normal MSCs are capable of down-regulating autophagy in activated T cells through the inhibition of respiratory mitochondrial biogenesis [20].

Genetic factors contributing to MSC dysfunction in SLE

Because MSCs from SLE patients still display impaired immunomodulatory function in vitro, it is assumed that the genetic background plays an essential role in MSC dysfunction. There is plenty of indirect evidence to imply genetic involvement in SLE MSC function (Table 1). Until recently, however, direct evidence of MSC genetic regulation in SLE has been reported in only a few studies.

Table 1 Factors that may be involved in the regulation of MSC function in SLE

Genes directly resulting in MSC abnormality

Olfactory 1/early B cell factor-associated zinc-finger protein (OAZ) is a transcription factor involved in bone morphogenetic protein (BMP)-induced signaling pathways [47]. As a candidate susceptibility gene of SLE, it plays a role in regulating anti-nuclear antibody production in SLE patients [48]. OAZ is over-expressed in MSCs from SLE patients [41]. Knockdown of the OAZ gene with small interfering RNAs restores the ability of MSCs to suppress B-cell proliferation and terminal differentiation, followed by a decrease in anti-nuclear antibody production. The effect of OAZ is mediated by the chemokine CCL2, as the level of CCL2 was increased after OAZ knockdown, while anti-CCL2 antibodies completely counteracted the effect of OAZ silencing.

Pre-B-cell leukemia homeobox 1 (Pbx1)-d, an isoform of Pbx1, is a dominant negative mutation located in murine NZM2410 lupus susceptibility locus Sle1a1, with a deletion of DNA binding exon 6 and Hox binding exon 7 [40]. The amino acid sequence of mouse Pbx1 is identical to that of the human protein, which preserves self-renewal of hematopoietic stem cells and blocks lineage-specific differentiation [49]. Evidence shows that Sle1a1 MSCs express higher levels of Pbx1-d than normal MSCs, resulting in the generation of activated auto-reactive CD4+ T cells and increased anti-chromatin IgG production by B cells [40]. Pbx1-d mutation is found more frequently in SLE patients than in normal controls [50], implying that Pbx1-d also works in humans to regulate MSC function.

p16INK4A, an inhibitor of cyclin-dependent kinase (CDK)4 and CDK6, is closely associated with senescence of MSCs [51]. The expression of p16INK4A has been shown to significantly increase in MSCs from SLE patients [52], which could account for the prominent senescence of lupus MSCs, as characterized by disordered cytoskeleton distribution and reduced immunoregulatory ability. Moreover, MSCs with p16INK4A knockdown express elevated levels of TGF-β, leading to an increased percentage of Treg cells when cultured with purified CD4+ T cells. Therefore, p16INK4A may cripple the function of SLE MSCs by both the induction of cell senescence and the inhibition of TGF-β secretion.

Indirect evidence of genetic involvement

PD-1 has been identified as a lupus susceptibility gene in European and Mexicans since 2002 [53]. The risk allele A of the intronic single nucleotide polymorphism (rs1033438163) alters a binding site for the runt-related transcription factor 1 (RUNX1) and inhibits the expression of PD-1. Decreased expression of PD-1 may impair MSC suppression of p38, ERK, and Akt phosphorylation signaling transduction in B-cell receptor activated B cells, leading to the loss of peripheral self-tolerance in B cells [23].

Low affinity immunoglobulin gamma Fc region receptor II-b (FcγRIIB), a low affinity receptor usually expressed on B cells and myeloid DCs, is another lupus susceptibility gene in different populations [54]. Recently, this gene has been proven to be expressed on MSCs [55]. FcγRIIB-deficient MSCs are less potent at suppressing dendritic cell maturation and the antigen-specific T-cell response, along with reduced expression of immunosuppressive factors such as PGE2 and cyclooxygenase-2.

STAT protein family mediates IFN signaling responses and plays a role in maintaining immune tolerance. Both STAT1 and STAT4 have been reported to be related to autoantibody production and renal involvement in either lupus mice or SLE patients [56,57,58]. A risk allele of STAT4 has been found to be over-expressed in SLE mesenchymal cells [59]. Meanwhile, down-regulation of STAT1 abrogates MSCs’ immunosuppressive capacity via the inhibition of IDO production, while over-expression of STAT1 significantly enhances IDO production in MSCs [60]. Thus, STAT proteins may serve as potential regulators of lupus MSCs.

Several cytokines and chemokines, such as IL-6, IL-7, IDO, and CCL2, have been found to be down-regulated in MSCs from SLE patients [6, 17, 39]. The impaired ability of MSCs to secrete these regulatory factors may be attributed to the genetic background of SLE patients and accordingly weakened MSC function. For example, IL-6 reduction restrains MSC-induced macrophage polarization, while lack of IDO leads to insufficiency of the down-regulation of T-cell proliferation.

Factors that are highly expressed in serum from SLE patients may also play a role in the regulation of MSC function. Evidence shows that TNF-α could increase MSC migration and invasion via activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway [61, 62]. Meanwhile, IFN-γ has been identified to stimulate IDO production in MSCs via the STAT1 signaling pathway [17], contributing to an enhanced immunosuppressive capacity of normal MSCs inhibiting the proliferation of lupus T cells. Recently, leptin and neutrophil-activating peptide 2 (NAP-2) have been shown to promote MSCs senescence through the activation of PI3K/Akt signaling pathway [63].

Conclusions

SLE MSCs display impaired immunosuppressive capacity to not only acquired immunity but also innate immunity. A lot of genetic factors may contribute to the dysfunction of MSCs. Delineation of the genetic mechanism may not only be helpful to clarify disease pathogenesis but also beneficial for the enhancement of MSC efficacy. With continued research, genetic engineering to modify MSCs with improved immunosuppressive functions will be achieved in the near future.

Abbreviations

AAM/M2:

Alternatively activated macrophages

BM:

Bone marrow

BMP:

Bone morphogenetic protein

CCL2:

Chemokine C-C motif ligand 2

CDK:

Cyclin-dependent kinase

DC:

Dendritic cell

FcγRIIB:

Gamma Fc region receptor II-b

IDO:

Indoleamine 2,3-dioxygenase

IFN:

Interferon

IL:

Interleukin

iNOS:

Inducible nitric oxide synthase

MMP:

Matrix metalloproteinase

MSC:

Mesenchymal stem cell

NAP-2:

Neutrophil-activating peptide 2

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cell

NK:

Natural killer

NO:

Nitric oxide

OAZ:

Olfactory 1/early B cell factor-associated zinc-finger protein

Pbx1:

Pre-B cell leukemia homeobox 1

PD-1:

Programmed death 1

PDL-1:

Programmed death ligand-1

PGE2:

Prostaglandin E2

RUNX1:

Runt-related transcription factor 1

SLE:

Systemic lupus erythematosus

STAT:

Signal transducer and activator of transcription

Tfh:

T follicular helper

TGF-β1:

Transforming growth factor beta1

TNF-α:

Tumor necrosis factor α

Treg:

T regulatory

References

  1. Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71–4.

    Article  PubMed  CAS  Google Scholar 

  2. Tavassoli M, Friedenstein A. Hemopoietic stromal microenvironment. Am J Hematol. 1983;15:195–203.

    Article  PubMed  CAS  Google Scholar 

  3. Stagg J. Immune regulation by mesenchymal stem cells: two sides to the coin. Tissue Antigens. 2007;69:1–9.

    Article  PubMed  CAS  Google Scholar 

  4. Wang D, Zhang H, Liang J, Li X, Feng X, Wang H, Hua B, Liu B, Lu L, Gilkeson GS, Silver RM, Chen W, Shi S, Sun L. Allogeneic mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus: 4 years of experience. Cell Transplant. 2013;22:2267–77.

    Article  PubMed  Google Scholar 

  5. Carrion F, Nova E, Ruiz C, Diaz F, Inostroza C, Rojo D, Monckeberg G, Figueroa FE. Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus. 2010;19:317–22.

    Article  PubMed  CAS  Google Scholar 

  6. Che N, Li X, Zhang L, Liu R, Chen H, Gao X, Shi S, Chen W, Sun L. Impaired B cell inhibition by lupus bone marrow mesenchymal stem cells is caused by reduced CCL2 expression. J Immunol. 2014;193:5306–14.

    Article  PubMed  CAS  Google Scholar 

  7. Liang J, Sun L. Mesenchymal stem cells transplantation for systemic lupus erythematosus. Int J Rheum Dis. 2015;18:164–71.

    Article  PubMed  Google Scholar 

  8. Sun LY, Akiyama K, Zhang HY, Yamaza T, Hou YY, Zhao SN, Xu T, Le A, Shi ST. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells. 2009;27:1421–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Xu JJ, Wang DD, Liu DY, Fan ZP, Zhang HY, Liu OS, Ding G, Gao RT, Zhang CM, Ding YZ, Bromberg JS, Chen WJ, Sun LY, Wang SL. Allogeneic mesenchymal stem cell treatment alleviates experimental and clinical Sjogren syndrome. Blood. 2012;120:3142–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Wang L, Wang L, Cong X, Liu G, Zhou J, Bai B, Li Y, Bai W, Li M, Ji H, Zhu D, Wu M, Liu Y. Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: safety and efficacy. Stem Cells Dev. 2013;22:3192–202.

    Article  PubMed  CAS  Google Scholar 

  11. Ribeiro A, Laranjeira P, Mendes S, Velada I, Leite C, Andrade P, Santos F, Henriques A, Graos M, Cardoso CM, Martinho A, Pais M, da Silva CL, Cabral J, Trindade H, Paiva A. Mesenchymal stem cells from umbilical cord matrix, adipose tissue and bone marrow exhibit different capability to suppress peripheral blood B, natural killer and T cells. Stem Cell Res Ther. 2013;4:125.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22.

    Article  PubMed  CAS  Google Scholar 

  13. Sato K, Ozaki K, Oh I, Meguro A, Hatanaka K, Nagai T, Muroi K, Ozawa K. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 2007;109:228–34.

    Article  PubMed  CAS  Google Scholar 

  14. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, Zhao RC, Shi Y. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2:141–50.

    Article  PubMed  CAS  Google Scholar 

  15. Zhang Z, Feng R, Niu L, Huang S, Deng W, Shi B, Yao G, Chen W, Tang X, Gao X, Feng X, Sun L. Human umbilical cord mesenchymal stem cells inhibit T follicular helper cell expansion through the activation of iNOS in lupus-prone B6.MRL-Faslpr mice. Cell Transplant. 2017;26:1031–42.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Meisel R, Zibert A, Laryea M, Gobel U, Daubener W, Dilloo D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 2004;103:4619–21.

    Article  PubMed  CAS  Google Scholar 

  17. Wang D, Feng X, Lu L, Konkel JE, Zhang H, Chen Z, Li X, Gao X, Lu L, Shi S, Chen W, Sun L. A CD8 T cell/indoleamine 2,3-dioxygenase axis is required for mesenchymal stem cell suppression of human systemic lupus erythematosus. Arthritis Rheumatol. 2014;66:2234–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. English K, Ryan JM, Tobin L, Murphy MJ, Barry FP, Mahon BP. 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.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Liu Q, Zheng H, Chen X, Peng Y, Huang W, Li X, Li G, Xia W, Sun Q, Xiang AP. Human mesenchymal stromal cells enhance the immunomodulatory function of CD8(+)CD28(−) regulatory T cells. Cell Mol Immunol. 2015;12:708–18.

    Article  PubMed  CAS  Google Scholar 

  20. Chen J, Wang Q, Feng X, Zhang Z, Geng L, Xu T, Wang D, Sun L. Umbilical cord-derived mesenchymal stem cells suppress autophagy of T cells in patients with systemic lupus erythematosus via transfer of mitochondria. Stem Cells Int. 2016;2016:4062789.

    PubMed  PubMed Central  Google Scholar 

  21. Fan L, Hu C, Chen J, Cen P, Wang J, Li L. Interaction between mesenchymal stem cells and B-cells. Int J Mol Sci. 2016;17:650.

    Article  PubMed Central  CAS  Google Scholar 

  22. Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, Santarlasci V, Mazzinghi B, Pizzolo G, Vinante F, Romagnani P, Maggi E, Romagnani S, Annunziato F. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells. 2006;24:386–98.

    Article  PubMed  CAS  Google Scholar 

  23. Schena F, Gambini C, Gregorio A, Mosconi M, Reverberi D, Gattorno M, Casazza S, Uccelli A, Moretta L, Martini A, Traggiai E. Interferon-gamma-dependent inhibition of B cell activation by bone marrow-derived mesenchymal stem cells in a murine model of systemic lupus erythematosus. Arthritis Rheum. 2010;62:2776–86.

    Article  PubMed  CAS  Google Scholar 

  24. Rafei M, Hsieh J, Fortier S, Li M, Yuan S, Birman E, Forner K, Boivin MN, Doody K, Tremblay M, Annabi B, Galipeau J. Mesenchymal stromal cell-derived CCL2 suppresses plasma cell immunoglobulin production via STAT3 inactivation and PAX5 induction. Blood. 2008;112:4991–8.

    Article  PubMed  CAS  Google Scholar 

  25. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, Robey PG, Leelahavanichkul K, Koller BH, Brown JM, Hu X, Jelinek I, Star RA, Mezey E. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15:42–9.

    Article  PubMed  CAS  Google Scholar 

  26. Zhang QZ, Su WR, Shi SH, Wilder-Smith P, Xiang AP, Wong A, Nguyen AL, Kwon CW, Le AD. Human gingiva-derived mesenchymal stem cells elicit polarization of m2 macrophages and enhance cutaneous wound healing. Stem Cells. 2010;28:1856–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Geng Y, Zhang L, Fu B, Zhang J, Hong Q, Hu J, Li D, Luo C, Cui S, Zhu F, Chen X. Mesenchymal stem cells ameliorate rhabdomyolysis-induced acute kidney injury via the activation of M2 macrophages. Stem Cell Res Ther. 2014;5:80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Wang C, Chen J, Sun L, Liu Y. TGF-beta signaling-dependent alleviation of dextran sulfate sodium-induced colitis by mesenchymal stem cell transplantation. Mol Biol Rep. 2014;41:4977–83.

    Article  PubMed  CAS  Google Scholar 

  29. Deng W, Chen W, Zhang Z, Huang S, Kong W, Sun Y, Tang X, Yao G, Feng X, Chen W, Sun L. Mesenchymal stem cells promote CD206 expression and phagocytic activity of macrophages through IL-6 in systemic lupus erythematosus. Clin Immunol. 2015;161:209–16.

    Article  PubMed  CAS  Google Scholar 

  30. Spaggiari GM, Abdelrazik H, Becchetti F, Moretta L. MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: central role of MSC-derived prostaglandin E2. Blood. 2009;113:6576–83.

    Article  PubMed  CAS  Google Scholar 

  31. Wehner R, Wehrum D, Bornhauser M, Zhao S, Schakel K, Bachmann MP, Platzbecker U, Ehninger G, Rieber EP, Schmitz M. Mesenchymal stem cells efficiently inhibit the proinflammatory properties of 6-sulfo LacNAc dendritic cells. Haematologica. 2009;94:1151–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Deng Y, Yi S, Wang G, Cheng J, Zhang Y, Chen W, Tai Y, Chen S, Chen G, Liu W, Zhang Q, Yang Y. Umbilical cord-derived mesenchymal stem cells instruct dendritic cells to acquire tolerogenic phenotypes through the IL-6-mediated upregulation of SOCS1. Stem Cells Dev. 2014;23:2080–92.

    Article  PubMed  CAS  Google Scholar 

  33. Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood. 2008;111:1327–33.

    Article  PubMed  CAS  Google Scholar 

  34. Shi D, Li X, Chen H, Che N, Zhou S, Lu Z, Shi S, Sun L. High level of reactive oxygen species impaired mesenchymal stem cell migration via overpolymerization of F-actin cytoskeleton in systemic lupus erythematosus. Pathol Biol (Paris). 2014;62:382–90.

    Article  CAS  Google Scholar 

  35. Li X, Liu L, Meng D, Wang D, Zhang J, Shi D, Liu H, Xu H, Lu L, Sun L. Enhanced apoptosis and senescence of bone-marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Stem Cells Dev. 2012;21:2387–94.

    Article  PubMed  CAS  Google Scholar 

  36. Geng L, Li X, Feng X, Zhang J, Wang D, Chen J, Liu R, Chen H, Sun L. Association of TNF-alpha with impaired migration capacity of mesenchymal stem cells in patients with systemic lupus erythematosus. J Immunol Res. 2014;2014:169082.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Tang Y, Ma X, Zhang H, Gu Z, Hou Y, Gilkeson GS, Lu L, Zeng X, Sun L. Gene expression profile reveals abnormalities of multiple signaling pathways in mesenchymal stem cell derived from patients with systemic lupus erythematosus. Clin Dev Immunol. 2012;2012:826182.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Becker-Merok A, Eilertsen GO, Nossent JC. Levels of transforming growth factor-beta are low in systemic lupus erythematosus patients with active disease. J Rheumatol. 2010;37:2039–45.

    Article  PubMed  CAS  Google Scholar 

  39. Sun LY, Zhang HY, Feng XB, Hou YY, Lu LW, Fan LM. Abnormality of bone marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Lupus. 2007;16:121–8.

    Article  PubMed  CAS  Google Scholar 

  40. Lu S, Zeumer L, Sorensen H, Yang H, Ng Y, Yu F, Riva A, Croker B, Wallet S, Morel L. The murine Pbx1-d lupus susceptibility allele accelerates mesenchymal stem cell differentiation and impairs their immunosuppressive function. J Immunol. 2015;194:43–55.

    Article  PubMed  CAS  Google Scholar 

  41. Feng X, Che N, Liu Y, Chen H, Wang D, Li X, Chen W, Ma X, Hua B, Gao X, Tsao BP, Sun L. Restored immunosuppressive effect of mesenchymal stem cells on B cells after olfactory 1/early B cell factor-associated zinc-finger protein down-regulation in patients with systemic lupus erythematosus. Arthritis Rheumatol. 2014;66:3413–23.

    Article  PubMed  CAS  Google Scholar 

  42. Lu L, Wang DD, Li X, Zeng XF, Sun LY. Mechanism of umbilical cord mesenchymal stem cells in the up-regulation of regulatory T cells by transforming growth factor beta1 in systemic lupus erythematosus. Zhonghua Yi Xue Za Zhi. 2013;93:980–3.

    PubMed  CAS  Google Scholar 

  43. Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141–51.

    Article  PubMed  CAS  Google Scholar 

  44. Katsiari CG, Liossis SN, Sfikakis PP. The pathophysiologic role of monocytes and macrophages in systemic lupus erythematosus: a reappraisal. Semin Arthritis Rheum. 2010;39:491–503.

    Article  PubMed  CAS  Google Scholar 

  45. Alessandri C, Barbati C, Vacirca D, Piscopo P, Confaloni A, Sanchez M, Maselli A, Colasanti T, Conti F, Truglia S, Perl A, Valesini G, Malorni W, Ortona E, Pierdominici M. T lymphocytes from patients with systemic lupus erythematosus are resistant to induction of autophagy. FASEB J. 2012;26:4722–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Gergely P Jr, Grossman C, Niland B, Puskas F, Neupane H, Allam F, Banki K, Phillips PE, Perl A. Mitochondrial hyperpolarization and ATP depletion in patients with systemic lupus erythematosus. Arthritis Rheum. 2002;46:175–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Hata A, Seoane J, Lagna G, Montalvo E, Hemmati-Brivanlou A, Massague J. OAZ uses distinct DNA- and protein-binding zinc fingers in separate BMP-Smad and Olf signaling pathways. Cell. 2000;100:229–40.

    Article  PubMed  CAS  Google Scholar 

  48. Feng X, Li R, Huang J, Zhang H, Zhu L, Hua B, Tsao BP, Sun L. Olf1/EBF associated zinc finger protein interfered with antinuclear antibody production in patients with systemic lupus erythematosus. Arthritis Res Ther. 2010;12:R59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Ficara F, Murphy MJ, Lin M, Cleary ML. Pbx1 regulates self-renewal of long-term hematopoietic stem cells by maintaining their quiescence. Cell Stem Cell. 2008;2:484–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Cuda CM, Li S, Liang S, Yin Y, Potula HH, Xu Z, Sengupta M, Chen Y, Butfiloski E, Baker H, Chang LJ, Dozmorov I, Sobel ES, Morel L. Pre-B cell leukemia homeobox 1 is associated with lupus susceptibility in mice and humans. J Immunol. 2012;188:604–14.

    Article  PubMed  CAS  Google Scholar 

  51. Shibata KR, Aoyama T, Shima Y, Fukiage K, Otsuka S, Furu M, Kohno Y, Ito K, Fujibayashi S, Neo M, Nakayama T, Nakamura T, Toguchida J. Expression of the p16INK4A gene is associated closely with senescence of human mesenchymal stem cells and is potentially silenced by DNA methylation during in vitro expansion. Stem Cells. 2007;25:2371–82.

    Article  PubMed  CAS  Google Scholar 

  52. Gu Z, Cao X, Jiang J, Li L, Da Z, Liu H, Cheng C. Upregulation of p16INK4A promotes cellular senescence of bone marrow-derived mesenchymal stem cells from systemic lupus erythematosus patients. Cell Signal. 2012;24:2307–14.

    Article  PubMed  CAS  Google Scholar 

  53. Prokunina L, Castillejo-Lopez C, Oberg F, Gunnarsson I, Berg L, Magnusson V, Brookes AJ, Tentler D, Kristjansdottir H, Grondal G, Bolstad AI, Svenungsson E, Lundberg I, Sturfelt G, Jonssen A, Truedsson L, Lima G, Alcocer-Varela J, Jonsson R, Gyllensten UB, Harley JB, Alarcon-Segovia D, Steinsson K, Alarcon-Riquelme ME. A regulatory polymorphism in PDCD1 is associated with susceptibility to systemic lupus erythematosus in humans. Nat Genet. 2002;32:666–9.

    Article  PubMed  CAS  Google Scholar 

  54. Jeon JY, Kim KY, Kim BS, Jung JY, Kim HA, Suh CH. FcgammaRIIB gene polymorphisms are associated with disease risk and clinical manifestations of systemic lupus erythematosus in Koreans. Tohoku J Exp Med. 2015;236:185–91.

    Article  PubMed  CAS  Google Scholar 

  55. Zhu T, Chen R, Li Z, Tian J, Deng C, Zhang X, Zhang K, Tong L, Yu Y, Bai C. Functional role of FcgammaRIIB in the regulation of mesenchymal stem cell function. Int J Med Sci. 2016;13:154–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Karonitsch T, Feierl E, Steiner CW, Dalwigk K, Korb A, Binder N, Rapp A, Steiner G, Scheinecker C, Smolen J, Aringer M. Activation of the interferon-gamma signaling pathway in systemic lupus erythematosus peripheral blood mononuclear cells. Arthritis Rheum. 2009;60:1463–71.

    Article  PubMed  Google Scholar 

  57. Liang Y, Xu WD, Yang XK, Fang XY, Liu YY, Ni J, Qiu LJ, Hui P, Cen H, Leng RX, Pan HF, Ye DQ. Association of signaling transducers and activators of transcription 1 and systemic lupus erythematosus. Autoimmunity. 2014;47:141–5.

    Article  PubMed  CAS  Google Scholar 

  58. Xu Z, Duan B, Croker BP, Morel L. STAT4 deficiency reduces autoantibody production and glomerulonephritis in a mouse model of lupus. Clin Immunol. 2006;120:189–98.

    Article  PubMed  CAS  Google Scholar 

  59. Sigurdsson S, Nordmark G, Garnier S, Grundberg E, Kwan T, Nilsson O, Eloranta ML, Gunnarsson I, Svenungsson E, Sturfelt G, Bengtsson AA, Jonsen A, Truedsson L, Rantapaa-Dahlqvist S, Eriksson C, Alm G, Goring HH, Pastinen T, Syvanen AC, Ronnblom L. A risk haplotype of STAT4 for systemic lupus erythematosus is over-expressed, correlates with anti-dsDNA and shows additive effects with two risk alleles of IRF5. Hum Mol Genet. 2008;17:2868–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Mounayar M, Kefaloyianni E, Smith B, Solhjou Z, Maarouf OH, Azzi J, Chabtini L, Fiorina P, Kraus M, Briddell R, Fodor W, Herrlich A, Abdi R. PI3kalpha and STAT1 interplay regulates human mesenchymal stem cell immune polarization. Stem Cells. 2015;33:1892–901.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Bocker W, Docheva D, Prall WC, Egea V, Pappou E, Rossmann O, Popov C, Mutschler W, Ries C, Schieker M. IKK-2 is required for TNF-alpha-induced invasion and proliferation of human mesenchymal stem cells. J Mol Med (Berl). 2008;86:1183–92.

    Article  CAS  Google Scholar 

  62. Lu ZY, Chen WC, Li YH, Li L, Zhang H, Pang Y, Xiao ZF, Xiao HW, Xiao Y. TNF-alpha enhances vascular cell adhesion molecule-1 expression in human bone marrow mesenchymal stem cells via the NF-kappaB, ERK and JNK signaling pathways. Mol Med Rep. 2016;14:643–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Chen H, Shi B, Feng X, Kong W, Chen W, Geng L, Chen J, Liu R, Li X, Chen W, Gao X, Sun L. Leptin and neutrophil-activating peptide 2 promote mesenchymal stem cell senescence through activation of the phosphatidylinositol 3-kinase/Akt pathway in patients with systemic lupus erythematosus. Arthritis Rheumatol. 2015;67:2383–93.

    Article  PubMed  CAS  Google Scholar 

  64. Sioud M, Mobergslien A, Boudabous A, Floisand Y. Evidence for the involvement of galectin-3 in mesenchymal stem cell suppression of allogeneic T-cell proliferation. Scand J Immunol. 2010;71:267–74.

    Article  PubMed  CAS  Google Scholar 

Download references

Funding

This work is supported by the National Natural Science Foundation of China (81771745) and Jiangsu Provincial Medical Talent Program (ZDRCA2016059).

Author information

Authors and Affiliations

Authors

Contributions

YZ drafted the manuscript and was a major contributor in writing the manuscript. XF revised the genetic factors part. Both of authors read and approved the final manuscript.

Corresponding author

Correspondence to Xuebing Feng.

Ethics declarations

Competing interests

The authors declare that no competing interests exist.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, Y., Feng, X. Genetic contribution to mesenchymal stem cell dysfunction in systemic lupus erythematosus. Stem Cell Res Ther 9, 149 (2018). https://doi.org/10.1186/s13287-018-0898-x

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/s13287-018-0898-x

Keywords