review article

The Role of Nitric Oxide in Stem/Progenitor Cells

Yuqi Cui1,2, Haoran Bu3, Lianqun Cui1*

1Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan Shandong, China

2Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA

3Department of Emergency, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China

*Corresponding author: Lianqun Cui, Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan Shandong, 324 Jingwuweiqi Road, Jinan, Shandong 250021, China. Tel: +86 53168777915; Email: csdslyy@sina.com

Received Date: 19 September, 2017; Accepted Date: 25 September, 2017; Published Date: 05 October, 2017

Citation: Cui Y, Bu H, Cui L (2017) The Role of Nitric Oxide in Stem/Progenitor Cells. J Pharma Pharma Sci 02: 141. DOI: 10.29011/2574-7711.100041

1. Introduction

NO, which was first discovered as Endothelium-Derived Relaxing Factors (EDRF) in cardiovascular system, has been established as a diffusible universal messenger that mediates cell-cell communication throughout the body and regulates different physiological and pathological processes in many tissues [1-7]. It works mainly through activation of its target receptor, the enzyme soluble Guanylate Cyclase (sGC), which, when activated, produces the second messenger cyclic-Guanosine Monophosphate (cGMP). Interestingly, a functional NO-cGMP signaling system that involves in development and early differentiation of Embryonic Stem Cells (ESCs) can be evolutionarily conserved between vertebrates and invertebrates [8]. In addition, NO, as a short-lived free radical gas is synthesized from L-arginine by a family of enzymes known as NO synthases (NOS) [9]. Three NOS isozymes encoded by three separate genes, including the Ca21/calmodulin-dependent and constitutively expressed neuronal NOS (nNOS), endothelial NOS (eNOS) enzymes, and a calmodulin-independent cytokine-inducible NOS (iNOS) enzyme found in various cell types [10]. A small amount of NO, produced by the constitutive NOS in response to increase in intracellular calcium, play a crucial role in numerous physiological functions, including neurotransmission [11], vascular tone [12] and platelet aggregation [13], whereas the large amounts, generated by iNOS, are implicated in pathological functions such as cytotoxicity of activated macrophages [14]. Recently, experimental evidence has been presented that not only can stem cells produce NOS, but its production, exogenous and endogenous NO, can also affect the proliferation, mobilization, and differentiation of different stem cells.

2. Role of NO in ESCs

ESCs are pluripotent stem cells derived from the inner cell mass of the blastocyst, an early-stage embryo. Krumenacker et al [15-17] have examined the expression of various subunits of sGC alpha (1), alpha (2), beta (1), beta (2), NOS, MLC2 (cardiac marker) and a cardiac-specific transcription factor (Nkx2.5) in human Embryonic Stem (hES) cells (H-9 cells) and differentiated cells subjected to differentiation in cell suspension using Embryoid Body (EB) formation. Their results clearly demonstrate the role of NO signaling components in differentiation events or physiological processes of human ES or ES cell-derived cardio myocytes. In addition, cGMP analysis in undifferentiated stem cells revealed a lack of stimulation with NO donors. Differentiated cells however, acquired the ability to be stimulated by NO donors. Although 3-(4-amino-5-cyclopropylpyrimidin-2-yl)-1-(2-fluorobenzyl)-1H-pyrazolo [3,4-b] pyridine (BAY 41-2272) alone was able to stimulate cGMP accumulation, the combination of NO donors and BAY 41-2272 stimulated cGMP levels more than either of the agents separately. These studies demonstrate that cGMP-mediated NO signaling plays an important role in the differentiation of ES cells into myocardial cells. Additionally, they also found nNOS and eNOS are detected in undifferentiated mouse ES cells while iNOS were very low or undetectable. However, although analysis of sGC activity in cell lysates derived from undifferentiated ES cells revealed that NO could not stimulate cGMP, lysates from differentiated EB outgrowths produced abundant cGMP levels after NO stimulation. Furthermore, purification of ES-cell derived Cardio Myocytes (CM) revealed that mRNA expression of all the NOS isoforms was very low to absent while sGCα1 and β1 subunit mRNAs were abundant and sGC-mediated cGMP production was apparent in this population of cells. These data suggest that cGMP-mediated NO signaling may play a minor role, if any, in undifferentiated ES cells but could be involved in the early differentiation events or physiological processes of ES cells or ES cell-derived lineages. Moreover, Huang et al. [18] suggests that NOS elements are present in Endothelial Cells (ECs) but inactive until later stages of differentiation, during which NOS inhibition reduces expression of EC markers and impairs angiogenic function. Further researches has been reported by Mora-Castilla S et al. [19] who indicate that exposure to 0.5 mM DETA-NO induces early differentiation events of cells with acquisition of epithelial morphology and expression of markers of definitive endoderm, such as FoxA2, Gata4, Hfn1-beta and Sox 17.

3.  Role of NO in EPC

EPCs are a controversial and hypothetical population of rare cells believed to circulate in the blood with the ability to differentiate into endothelial cells, the cells that make up the lining of blood vessels. The process by which blood vessels are born de novo from endothelial progenitor cells is known as vasculogenesis which involves in NO that can stimulate endothelial cell proliferation, survival and motility, and enhances matrix invasion and tubulogenesis with the help of the pro-angiogenic activity of Growth Factors such as VEGF, transforming growth factor-β and FGF [20-23]. Downstream mediators of NO, including cAMP- and cGMP-dependent protein kinases [PKA (Protein Kinase A) and PKG (Protein Kinase G)], Rho GTPases and ROS (Reactive Oxygen Species), are likely to play a part. Recent studies have showed that the activity of Rho GTPases, key regulators of endothelial cell motility and angiogenesis is modulated by altering the metabolism of Asymmetric Dimethyl Arginine (ADMA) which is a cardiovascular risk factor, an endogenous inhibitor of NOS, increased when abnormal angiogenesis in cardiovascular disorder happened, and is metabolized by Dimethylarginine Dimethyl Amino Hydrolases (DDAHs) in vivo and in vitro. Fiedler et al. [24] believed that the ADMA/DDAH pathway is to regulate angiogenesis by influencing NO bioavailability [25]. Consistent with the role of NO in the ADMA/DDAH pathway, DDAH I gene deletion in mice leads to inhibition of angiogenic responses, similar to that seen in eNOS-knockout mice [22,26-28]. Additionally, NO also modulates gene expression of factors that promote angiogenesis, such as αvβ3 integrin, and suppresses the production of antiangiogenic factors such as angiostatin, the degradation product of plasminogen [20]. And Shen et al. [29] indicate that suppressed NO production from EPCs was involved in the glycation end products (AGE)-induced apoptosis, which is in part mediated by Mitogen-Activated Protein Kinases (MAPKs) signaling.

MSCs are multipotent stem cells that can differentiate into a variety of cell types include osteoblasts, chondrocytes and adipocytes. Tatsumi R et al. [30] demonstrated that the quiescent satellite cells of resident myogenic stem cells which are from MSCs are activated to enter the cell proliferation cycle, divide, differentiate, and fuse with the adjacent muscle fiber, and are responsible for regeneration and work-induced hypertrophy of muscle fibers by activation mechanism which is a cascade of events including calcium ion influx, calcium-calmodulin formation, NO synthase activation, NO radical production when muscle is injured, exercised, overused or mechanically stretched. Therefore, MSCs have received special attention for cardiomyoplasty. Rebelatto et al. [31] report an investigation of the effects of two NO agents (SNAP and DEA/NO), able to activate both cGMP-dependent and independent pathways, on the cardio myogenic potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) and Adipose Tissue-Derived Stem Cells (ADSCs). They found that untreated (control) ADSCs and BM-MSCs expressed some muscle markers and NO-derived intermediates induce an increased expression of some cardiac function genes in BM-MSCs and ADSCs. And NO agents considerably increased the pro-angiogenic potential mostly of BM-MSCs as determined by VEGF mRNA levels. Additionally, Mookerjee et al. [32] investigated the signaling mechanisms by Human gene-2 (H2) relaxin which can inhibit renal my fibroblast differentiation by interfering with TGF-beta1/Smad2 signaling regulates myofibroblast differentiation in vitro by examining its effects on mixed populations of fibroblasts and myofibroblasts propagated from injured rat kidneys. Inhibition of nNOS, NO, and cGMP significantly blocked the inhibitory effects of relaxin on alpha-SMA and Smad2 phosphorylation, while the NO inhibitor, L-Nitro Arginine Methyl Ester (hydrochloride) (L-NAME) significantly blocked the inhibitory actions of relaxin on collagen concentration in vivo. Moreover, Bironaite et al. [33] clearly demonstrate sustained activation of MAPKs which actively participate in the regulation of cell survival and of proapoptotic signals in myogenic stem cells after exposure to the NO inducer, NOC-18. Inhibition of MAPKs phosphorylation by specific inhibitors revealed the anti-apoptotic role of MAPKs in myogenic stem cells. On the other hand, Kraft DCE et al. [34] indicate that Pulsating fluid flow (PFF) stimulated NO production within 5 min by human dental Pulp-Derived Mesenchymal Stem Cells (PDSCs) portraying mature (PDSC-mature) but not by PDSC immature. The rapid stimulation of NO production by PFF in PDSCs is probably a result of the activity of eNOS, but not iNOS, since unlike iNOS, eNOS is constitutively expressed in bone cells and dental pulp cells. Additionally, NO produced by eNOS is primarily regulated by Ca2+ fluxes and subsequent binding of calmodulin, and eNOS only produces NO for minutes after stimulation [35-37] demonstrate that NO-induced osteogenic differentiation through Heme Oxygenase-1 (HO-1) may be an important mediator of periodontal regeneration or bone tissue engineering.

5. Role of NO in NSCs

NSCs can be propagated for extended periods of time and differentiated into both neuronal and glia cells. Tegenge et al. [38] indicate that NO plays a role in the development of the human nervous system. They used a model of human Neuronal Precursor Cells (NPCs) from a well-characterized teratocarcinoma cell line (NT2). Their results from the differentiating NT2 model neurons point towards a vital role of the NO/cGMP/PKG signaling cascade as positive regulator of cell migration in the developing human brain. And Yoneyama et al. [39] also suggest that NO and endogenous ROS are essential for the proliferation of embryonic NSCs and NPCs. However, NO can also cause apoptosis of Neural Progenitor Cells (NPCs). Hung et al. [40] studied the role of p53 in the NO-induced apoptosis was examined in an in vitro model of NPCs. Their results suggest a central role of p53 in the NO-induced apoptotic pathway in NPCs, which may hence provide new insights into the regulation of cell death in NPCs that respond to overproduction of NO in injured brain.

6. Current NO Research on Various Diseases by Stem Cells

6.1. Gene Therapy Techniques for Pulmonary Hypertension

 NO synthesized by eNOS is important in regulating vascular resistance and in vascular remodeling in the lung. NO deficiency due to endothelial dysfunction plays an important role in the pathogenesis of Pulmonary Hypertension (PH) which is a serious, often fatal disease characterized by remodeling of the pulmonary vascular bed, increase pulmonary arterial pressure, and right heart failure. Deng et al. [41] describe the use of two gene transfer techniques, i.e., adenoviral gene transfers of eNOS and eNOS gene-modified rat marrow stromal cells, for eNOS gene delivery to the lung of laboratory animals for the treatment of PH. Therefore, local eNOS gene delivery to the lung is a promising approach for the treatment of PH and Adenoviral-mediated in vivo gene therapy and adult stem cell-based ex vivo gene therapy are two attractive current gene therapies for the treatment of cardiovascular and pulmonary diseases.
6.2. Gene Therapy Techniques for Fibro Sarcoma

Emerging evidence suggests that MSC are able to migrate to sites of tissue injury and have immunosuppressive properties that may be useful in targeted gene therapy for sustained specific tissue engraftment. Xiang et al. [42] observed that xenogenic MSC selectively migrated to the tumor site, proliferated and expressed the exogenous gene in subcutaneous fibro sarcoma transplants and no MSC distribution was detected in other organs, such as the liver, spleen, colon and kidney. They further showed that the FGF2/FGFR pathways may play a role in the directional movement of MSC to the Rif-1 fibro sarcoma and they performed in vitro co-culture and in vivo tumor growth analysis, showing that MSC did not affect the proliferation of Rif-1 cells and fibro sarcoma growth compared with an untreated control group. Finally, they demonstrated that the xenogenic MSC stably expressing iNOS protein transferred by a lentivirus-based system had a significant inhibitory effect on the growth of Rif-1 tumors compared with MSC alone and the non-treatment control group. Therefore, iNOS delivered by genetically modified iNOS-MSC showed a significant anti-tumor effect both in vitro and in vivo. MSC may be used as a target gene delivery vehicle for the treatment of fibro sarcoma and other tumors.

6.3. Perivascular NO Involved in Stem-Like Character in PDGF-Induced Glioma Cells

eNOS expression is elevated in human glioblastomas and correlated with increased tumor growth and aggressive character. Charles et al. [43] investigated the potential role of NO activity in the Perivascular Niche (PVN) using a genetic engineered mouse model of PDGF-induced gliomas. eNOS expression is highly elevated in tumor vascular endothelium adjacent to perivascular glioma cells expressing Nestin, Notch, and the NO receptor, sGC. In addition, the NO/cGMP/PKG pathway drives Notch signaling in PDGF-induced gliomas in vitro, and induces the side population phenotype in primary glioma cell cultures. NO also increases neurosphere forming capacity of PDGF-driven glioma primary cultures, and enhances their tumorigenic capacity in vivo. Loss of NO activity in these tumors suppresses Notch signaling in vivo and prolongs survival of mice. This mechanism is conserved in human PDGFR amplified gliomas. The NO/cGMP/PKG pathway's promotion of stem cell-like character in the tumor PVN may identify therapeutic targets for this subset of gliomas.

 6.4. NO Involved in The Therapy for Cardiovascular Disease

 Recent studies have reported a marked impairment in the number and functions of EPCs in patients with Coronary Artery Disease (CAD). LiN [44] found that eNOS in the host myocardium promotes MSC migration to the ischemic myocardium and improves cardiac function through cGMP-dependent increases in cell-derived factor-1alpha (SDF-1alpha) expression. Furthermore, Kaur et al. [45] conclude that eNOS gene transfection is a valuable approach to augment angiogenic properties of ex vivo expanded EPCs and eNOS-modified EPCs may offer significant advantages than EPCs alone in terms of their clinical use in patients with myocardial ischemia. Moreover, Spallotta et al. [46] found that NO-treated ES injected into the cardiac left ventricle selectively localized in the ischemic hind limb and contributed to the regeneration of muscular and vascular structures. These findings establish a key role for NO in therapy of cardiovascular diseases.

6.5. NO Involved in The Immunosuppression

MSCs hold great promise for treating immune disorders because of their immunoregulatory capacity and the mechanism of MSC-mediated immunosuppression varies among different species. Immunosuppression by human- or monkey-derived MSCs is mediated by Indole amine 2,3-Dioxygenase (IDO), whereas mouse MSCs utilize NO, under the same culture conditions. When the expression of IDO and iNOS were examined in human and mouse MSCs after stimulation with their respective inflammatory cytokines, Ren et al. [47] found that human MSCs expressed extremely high levels of IDO, and very low levels of iNOS, whereas mouse MSCs expressed abundant iNOS and very little IDO. However, immunosuppression by human MSCs was not intrinsic, but was induced by inflammatory cytokines and was chemokine-dependent, as it is in mouse. Further studies have reported that NSCs may exert direct anti-inflammatory activity. This action has been attributed, in part, to T-cell suppression. Wang et al. [48] indicate that NSCs appear to suppress T-cells, at least in part, by NO and Prostaglandin E2 (PGE2) production which, in turn, would account for the well-documented reduction of central nervous system immunopathology by transplanted NSCs. These findings provide critical information about the immunosuppression of MSCs and for better application of MSCs in treating immune disorders.
 7. Conclusions

In summary, the downstream mediators of NO and NO itself are likely to exert the function of modulation in the process of EPCs differentiation. Although scarcely can NO influence the undifferentiated ESCs, it can be dramatically involved in the early differentiation events or physiological processes of ES cells or ES cell-derived lineages. Nevertheless, overproduction of NO may induce the apoptosis of NPCs. Therefore, NO plays an important role in physiological and pathological processes of stem cells and we can utilize these characters in the treatment of various diseases by various methods like gene transfer techniques, stem cell transplantation et al.
 8. Acknowledgement

 This work was supported by The National Nature Science Foundation of China to Yuqi Cui (81600222) and Shandong Provincial Nature Science Foundation, China to Lianqun Cui (ZR2016HM22).



  1. Feelisch M, Te Poel M, Zamora R, Deussen A, Moncada S (1994) Understanding the controversy over the identity of EDRF. Nature 368: 62-65.
  2. Hobbs AJ, Ignarro LJ (1996) Nitric oxide-cyclic GMP signal transduction system. Methods Enzymol 269: 134-148.
  3. Keifer HM, Kalak N, Tshudin K, Fingerle J, Van der Kooij M, et al. (2002) Inhibition of NO biosynthesis, but not elevated blood pressure, reduces angiogenesis in rat models of secondary hypertension. Blood Press 11: 116--124.
  4. MacMicking J, Xie QW, Nathan C (1997) Nitric oxide and macrophage function. Annu Rev Immunol 15: 323-350.
  5. Marletta MA (1993) Nitric oxide synthase: function and mechanism. J Biol Chem 268: 12231-12234.
  6. McGeer PL, Kawamata T, Walker DG, Akiyama H, Tooyama I, et al. (1993) Microglia in degenerative neurological disease. Glia 7: 84-92.
  7. Stamler JS, Jia L, Eu JP, McMahon TJ, Demchenko IT, et al. (1997) Blood flow regulation by S-nitrosohemoglobin in the physiological oxygen gradient. Science 276: 2034-2037.
  8. Colasanti M, Mazzone V, Mancinelli L, Leone S, Venturini G (2009) Involvement of nitric oxide in the head regeneration of Hydra vulgaris. Nitric oxide : biology and chemistry/official journal of the Nitric Oxide Society 21: 164-170.
  9. Lajoix AD, Reggio H, Chardes T, Peraldi-Roux S, Tribillac F, et al. (2001) A neuronal isoform of nitric oxide synthase expressed in pancreatic beta-cells controls insulin secretion. Diabetes 50: 1311-1323.
  10. Moncada S, Palmer RM, Higgs EA (1991) Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev 43: 109-142.
  11. Bult H, Boeckxstaens GE, Pelckmans PA, Jordaens FH, Van Maercke YM, et al. (1990) Nitric oxide as an inhibitory non-adrenergic non-cholinergic neurotransmitter. Nature 345: 346-347.
  12. Palmer RM, Rees DD, Ashton DS, Moncada S (1988) L-arginine is the physiological precursor for the formation of nitric oxide in endothelium-dependent relaxation. Biochemical and biophysical research communications 153: 1251-1256.
  13. Radomski MW, Palmer RM, Moncada S (1990) An L-arginine/nitric oxide pathway present in human platelets regulates aggregation. Proc Natl Acad Sci USA 87: 5193-5197.
  14. Hibbs JB, Taintor RR, Vavrin Z, Rachlin EM (1988) Nitric oxide: a cytotoxic activated macrophage effector molecule. Biochemical and biophysical research communications 157: 87-94.
  15. Krumenacker JS, Katsuki S, Kots A, Murad F (2006) Differential expression of genes involved in cGMP-dependent nitric oxide signaling in murine Embryonic Stem (ES) cells and ES cell-derived cardiomyocytes. Nitric Oxide 14: 1-11.
  16. Mujoo K, Krumenacker JS, Wada Y, Murad F (2006) Differential expression of nitric oxide signaling components in undifferentiated and differentiated human embryonic stem cells. Stem Cells Dev 15: 779-787.
  17. Mujoo K, Sharin VG, Bryan NS, Krumenacker JS, Sloan C, et al. (2008) Role of nitric oxide signaling components in differentiation of embryonic stem cells into myocardial cells. Proc Natl Acad Sci U S A 105: 18924-18929.
  18. Huang N, Fleissner F, Sun J, Cooke J (2010) Role of Nitric Oxide Signaling in Endothelial Differentiation of Embryonic Stem Cells. Stem Cells Dev 19: 1617-1627.
  19. Mora-Castilla S, Tejedo JR, Hmadcha A, Cahuana GM, Martin F, et al. (2010) Nitric oxide repression of Nanog promotes mouse embryonic stem cell differentiation. Cell Death Differ 17: 1025-1033.
  20. Cooke JP (2003) NO and angiogenesis. Atheroscler Suppl 4: 53-60.
  21. Goligorsky MS, Abedi H, Noiri E, Takhtajan A, Lense S, Romanov V, et al. (1999) Nitric oxide modulation of focal adhesions in endothelial cells. Am J Physiol 276: C1271-1281.
  22. Murohara T, Asahara T (2002) Nitric oxide and angiogenesis in cardiovascular disease. Antioxid Redox Signal 4: 825-831.
  23. Olsson A-K, Dimberg A, Kreuger J, Claesson-Welsh L (2006) VEGF receptor signalling-in control of vascular function. Nat Rev Mol Cell Biol 7: 359-371.
  24. Fiedler LR, Wojciak-Stothard B (2009) The DDAH/ADMA pathway in the control of endothelial cell migration and angiogenesis. Biochem Soc Trans 37: 1243-1247.
  25. Jacobi J, Sydow K, Von Degenfeld G, Zhang Y, Dayoub H,et al. (2005) Overexpression of dimethylarginine dimethylaminohydrolase reduces tissue asymmetric dimethylarginine levels and enhances angiogenesis. Circulation 111: 1431-1438.
  26. Achan V, Ho HK, Heeschen C, Stuehlinger M, Jang JJ, et al. (2005) ADMA regulates angiogenesis: genetic and metabolic evidence. Vasc Med 10: 7-14.
  27. Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz MA, et al. (1995) Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 377: 239-242.
  28. Wojciak-Stothard B, Torondel B, Tsang LYF, Fleming I, Fisslthaler B, et al. (2007) The ADMA/DDAH pathway is a critical regulator of endothelial cell motility. J Cell Sci 120: 929-942.
  29. Shen C, Li Q, Zhang YC, Ma G, Feng Y, et al. (2010) Advanced glycation endproducts increase EPC apoptosis and decrease nitric oxide release via MAPK pathways. Biomed Pharmacother 64: 35-43.
  30. Tatsumi R (2010) Mechano-biology of skeletal muscle hypertrophy and regeneration: Possible mechanism of stretch-induced activation of resident myogenic stem cells. Anim Sci J 81: 11-20.
  31. Rebelatto CK, Aguiar AM, Senegaglia AC, Aita CM, Hansen P, et al. (2009) Expression of cardiac function genes in adult stem cells is increased by treatment with nitric oxide agents. Biochemical and biophysical research communications 378: 456-461.
  32. Mookerjee I, Hewitson TD, Halls ML, Summers RJ, Mathai ML, et al. (2009) Relaxin inhibits renal myofibroblast differentiation via RXFP1, the nitric oxide pathway, and Smad2. Faseb J 23: 1219-1229.
  33. Bironaite D, Baltriukiene D, Uralova N, Stulpinas A, Bukelskiene V,et al. (2009) Role of MAP kinases in nitric oxide induced muscle-derived adult stem cell apoptosis. Cell Biol Int 33: 711-719.
  34. Kraft DC, Melsen B, Abdallah BM, Kassem M, Klein-Nulend J (2010) Mechanosensitivity of dental pulp stem cells is related to their osteogenic maturity. Eur J Oral Sci 118: 29-38.
  35. Di Nardo Di Maio F, Lohinai Z, D'Arcangelo C, De Fazio PE, Speranza L, et al. (2004) Nitric oxide synthase in healthy and inflamed human dental pulp. J Dent Res 83: 312-316.
  36. Klein-nulend JHM, Sterck JG, Macpherson H, Joldersma M, Ralston SH, et al. (1998) Nitric oxide response to shear stress by human bone cell cultures is endothelial nitric oxide synthase dependent. Biochem Biophys Res Commun 250: 108-114.
  37. Lee S-K, Choi H-I, Yang Y-S, Jeong G-S, Hwang J-H, et al. (2009) Nitric oxide modulates osteoblastic differentiation with heme oxygenase-1 via the mitogen activated protein kinase and nuclear factor-kappaB pathways in human periodontal ligament cells. Biol Pharm Bull 32: 1328-1334.
  38. Tegenge MA, Bicker G (2009) Nitric oxide and cGMP signal transduction positively regulates the motility of human neuronal precursor (NT2) cells. J Neurochem 110: 1828-1841.
  39. Yoneyama M, Kawada K, Gotoh Y, Shiba T, Ogita K (2009) Endogenous reactive oxygen species are essential for proliferation of neural stem/progenitor cells. Neurochem Int 56: 740-746.
  40. Hung AC, Porter AG (2009) p53 mediates nitric oxide-induced apoptosis in murine neural progenitor cells. Neurosci Lett 467: 241-246.
  41. Deng W, Bivalacqua TJ, Champion HC, Hellstrom WJ, Murthy SN, et al. (2010) Gene therapy techniques for the delivery of endothelial nitric oxide synthase to the lung for pulmonary hypertension. Methods Mol Biol 610: 309-321.
  42. Xiang J, Tang J, Song C, Yang Z, Hirst DG, et al. (2009) Mesenchymal stem cells as a gene therapy carrier for treatment of fibrosarcoma. Cytotherapy 11: 516-526.
  43. Charles N, Ozawa T, Squatrito M, Bleau AM, Brennan CW, et al. (2010) Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell 6: 141-152.
  44. Li N, Lu X, Zhao X, Xiang F-L, Xenocostas A, et al. (2009) Endothelial nitric oxide synthase promotes bone marrow stromal cell migration to the ischemic myocardium via upregulation of stromal cell-derived factor-1alpha. Stem cells (Dayton, Ohio) 27: 961-970.
  45. Kaur S, Kumar TR, Uruno A, Sugawara A, Jayakumar K, Kartha CC (2009) Genetic engineering with endothelial nitric oxide synthase improves functional properties of endothelial progenitor cells from patients with coronary artery disease: an in vitro study. Basic Res Cardiol 104: 739-749.
  46. Spallotta F, Rosati J, Straino S, Nanni S, Grasselli A, et al. (2010) Nitric oxide Determines Mesodermic Differentiation of Mouse Embryonic Stem Cells by Activating Class IIa Histone Deacetylases: Potential Therapeutic Implications in a Mouse Model of Hindlimb Ischemia. Stem Cells 28: 431-442.
  47. Ren G, Su J, Zhang L, Zhao X, Ling W, et al. (2009) Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells 27: 1954-1962.
  48. Wang L, Shi J, Van Ginkel FW, Lan L, Niemeyer G, et al. (2009) Neural stem/progenitor cells modulate immune responses by suppressing T lymphocytes with nitric oxide and prostaglandin E2. Exp Neurol 216:  177-183.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Pharmaceutical and Pharmacological Sciences

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131