Scientific References

At Anova IRM, we work by the principles of translational medicine. Following this and our ethical code, we only apply and recommend the use of stem cells if there is a clear scientific rational for the specific case and disease. A rational can come from guide lines, clinical trials, pre-clinical trials or a strong scientific rational - always weighted against risk and safety associated. This is a list of literature relevant for our work at Anova IRM. Literature research for the latest scientific literature is done in every patient case. Therefore, the list is neither complete nor reflects our entire library and working principles.

References and Literature - Stem Cell Secretome

  1. Konala, Vijay Bhaskar Reddy, et al. "The current landscape of the mesenchymal stromal cell secretome: a new paradigm for cell-free regeneration." Cytotherapy 18.1 (2016): 13-24.
  2. Lopez-Verrilli, M. A., et al. "Mesenchymal stem cell-derived exosomes from different sources selectively promote neuritic outgrowth." Neuroscience 320 (2016): 129-139.
  3. Kim, Hyun Ok, Seong-Mi Choi, and Han-Soo Kim. "Mesenchymal stem cell-derived secretome and microvesicles as a cell-free therapeutics for neurodegenerative disorders." Tissue Engineering and Regenerative Medicine 10.3 (2013): 93-101.
  4. Rani, Sweta, et al. "Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications." Molecular Therapy 23.5 (2015): 812-823.
  5. Zhang, Xiaoyan, et al. "Mesenchymal Stem Cell-Derived Extracellular Vesicles: Roles in Tumor Growth, Progression, and Drug Resistance." Stem Cells International 2017 (2017).
  6. omzikova, Marina O., and Albert A. Rizvanov. "Current Trends in Regenerative Medicine: From Cell to Cell-Free Therapy." BioNanoScience (2016): 1-6.
  7. Zhang, Bin, et al. "Focus on extracellular vesicles: Therapeutic potential of stem cell-derived extracellular Vesicles." International journal of molecular sciences 17.2 (2016): 174.
  8. Katsuda T. et al. (2013). Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Scientific reports, 3, 1197.
  9. Pusic A. D. et al. (2014). IFNγ-stimulated dendritic cell exosomes as a potential therapeutic for remyelination. Journal of neuroimmunology, 266(1), 12-23.
  10. Maumus M, Jorgensen C, Noël D (2013) Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: Role of secretome and exosomes. Biochimie 95:2229–2234. doi: 10.1016/j.biochi.2013.04.017
  11. Drago D, Cossetti C, Iraci N, et al (2013) Biochimie The stem cell secretome and its role in brain repair. Biochimie 95:2271–2285. doi: 10.1016/j.biochi.2013.06.020
  12. Sevivas N, Teixeira FG, Portugal R, et al (2016) Mesenchymal Stem Cell Secretome: A Potential Tool for the Prevention of Muscle Degenerative Changes Associated With Chronic Rotator Cuff Tears. Am J Sports Med. doi: 10.1177/0363546516657827
  13. Hs K (2016) Mesenchymal Stem Cells vs . Mesenchymal Stem Cell Secretome for Rheumatoid Arthritis Treatment. 1:1–2.
  14. Maumus M, Jorgensen C, Noël D (2013) Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: Role of secretome and exosomes. Biochimie 95:2229–2234. doi: 10.1016/j.biochi.2013.04.017
  15. Kapur SK, Katz AJ (2013) Biochimie Review of the adipose derived stem cell secretome. Biochimie 95:2222–2228. doi: 10.1016/j.biochi.2013.06.001
  16. Ranganath SH, Levy O, Inamdar MS, Karp JM (2012) Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell 10:244–258. doi: 10.1016/j.stem.2012.02.005
  17. Tran C, Damaser MS (2015) Stem cells as drug delivery methods: Application of stem cell secretome for regeneration. Adv Drug Deliv Rev 82:1–11. doi: 10.1016/j.addr.2014.10.007
  18. Zimmerlin L, Park TS, Zambidis ET, et al (2013) Mesenchymal stem cell secretome and regenerative therapy after cancer. Biochimie 95:2235–2245. doi: 10.1016/j.biochi.2013.05.010
  19. Calamia V, Lourido L, Fernandez-Puente P, et al (2012) Secretome analysis of chondroitin sulfate-treated chondrocytes reveals its anti-angiogenic, anti-inflammatory and anti-catabolic properties. Arthritis Res Ther 14:R202. doi: 10.1186/ar4040
  20. Ranganath SH, Levy O, Inamdar MS, Karp JM (2012) Review Harnessing the Mesenchymal Stem Cell Secretome for the Treatment of Cardiovascular Disease. Stem Cell 10:244–258. doi: 10.1016/j.stem.2012.02.005
  21. Teixeira FG, Carvalho MM, Sousa N, Salgado AJ (2013) Mesenchymal stem cells secretome: A new paradigm for central nervous system regeneration? Cell Mol Life Sci 70:3871–3882. doi: 10.1007/s00018-013-1290-8
  22. Kapur SK, Katz AJ (2013) Review of the adipose derived stem cell secretome. Biochimie 95:2222–2228. doi: 10.1016/j.biochi.2013.06.001
  23. Chang C-P, Chio C-C, Cheong C-U, et al (2013) Hypoxic preconditioning enhances the therapeutic potential of the secretome from cultured human mesenchymal stem cells in experimental traumatic brain injury. Clin Sci (Lond) 124:165–76. doi: 10.1042/CS20120226
  24. Salgado AJ, Sousa JC, Costa BM, et al (2015) Mesenchymal stem cells secretome as a modulator of the neurogenic niche: basic insights and therapeutic opportunities. Front Cell Neurosci 9:1–18. doi: 10.3389/fncel.2015.00249
  25. Ahmed NE-MB, Murakami M, Hirose Y, Nakashima M (2016) Therapeutic Potential of Dental Pulp Stem Cell Secretome for Alzheimer’s Disease Treatment: An In Vitro Study. Stem Cells Int 2016:8102478. doi: 10.1155/2016/8102478
  26. Bhaskar V, Konala R, Mamidi MK, et al (2016) The current landscape of the mesenchymal stromal cell secretome : A new paradigm for cell-free regeneration. Cytotherapy 18:13–24. doi: 10.1016/j.jcyt.2015.10.008
  27. Malda J, Boere J, van de Lest C, et al (2016) Extracellular vesicles - new tool for joint repair and regeneration - IN PRESS. Nat Rev Rheumatol 12:243–249. doi: 10.1038/nrrheum.2015.170
  28. Lener T, Gimona M, Aigner L, et al (2015) Applying extracellular vesicles based therapeutics in clinical trials Á an ISEV position paper. 1:1–31.
  29. Dostert G, Mesure B, Menu P, Velot É (2017) How Do Mesenchymal Stem Cells Influence or Are Influenced by Microenvironment through Extracellular Vesicles Communication ? 5:1–7. doi: 10.3389/fcell.2017.00006
  30. Joshi P, Benussi L, Furlan R, et al (2015) Extracellular vesicles in Alzheimer’s disease: Friends or foes? focus on Aβ-vesicle interaction. Int. J. Mol. Sci. 16:4800–4813.
  31. Gao T, Guo W, Chen M, et al (2016) Extracellular Vesicles and Autophagy in Osteoarthritis.
  32. Katsuda T, Ochiya T (2015) Molecular signatures of mesenchymal stem cell-derived extracellular vesicle-mediated tissue repair. Stem Cell Res Ther 6:212. doi: 10.1186/s13287-015-0214-y
  33. Lener T, Gioma M, Aigner L, et al (2015) Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position paper. J Extracell Vesicles 4:1–31. doi: 10.3402/jev.v4.30087
  34. Xu Y, Guo S, Wei C, et al (2016) The Comparison of Adipose Stem Cell and Placental Stem Cell in Secretion Characteristics and in Facial Antiaging.
  35. Buul GM Van, Villafuertes E, Bos PK, et al (2012) Mesenchymal stem cells secrete factors that inhibit in fl ammatory processes in short-term osteoarthritic synovium and cartilage explant culture. Osteoarthr Cartil 20:1186–1196. doi: 10.1016/j.joca.2012.06.003
  36. Baglio SR, Pegtel DM, Baldini N (2012) Mesenchymal stem cell secreted vesicles provide novel opportunities in ( stem ) cell-free therapy. 3:1–11. doi: 10.3389/fphys.2012.00359
  37. Anderson JD, Pham MT, Contreras Z, et al (2016) Mesenchymal stem cell-based therapy for ischemic stroke. Chinese Neurosurg J 2:36. doi: 10.1186/s41016-016-0053-4
  38. Biology C, Cell R, Eye N, Institutes N (2017) Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. 1273–1285.

References and Literature - Bone Marrow Concentrate 'BMC'

  1. He Y, He W, Qin G, Luo J, Xiao M. Transplantation KCNMA1 modified bone marrow-mesenchymal stem cell therapy for diabetes mellitus-induced erectile dysfunction. Andrologia. 2014;46(5):479-486. doi:10.1111/and.12104.
  2. Mathiasen AB, Qayyum AA, Jørgensen E, et al. Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: a randomized placebo-controlled trial ({MSC}-{HF} trial). Eur Heart J. 2015;36(27):1744-1753. doi:10.1093/eurheartj/ehv136.
  3. Mathiasen AB, Qayyum AA, Jørgensen E, et al. Interventional cardiology Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure?: a randomized placebo-controlled trial. 2015. doi:10.1093/eurheartj/ehv136.
  4. Liao H-T, Chen C-T. Osteogenic potential: Comparison between bone marrow and adipose-derived mesenchymal stem cells. World J Stem Cells. 2014;6(3):288-295. doi:10.4252/wjsc.v6.i3.288.
  5. Terai S, Ishikawa T, Omori K, et al. Improved liver function in patients with liver cirrhosis after autologous bone marrow cell infusion therapy. Stem Cells. 2006;24(10):2292-2298. doi:10.1634/stemcells.2005-0542.
  6. 2015_Cao_Spine-Journal_Bone-marrow-mesenchymal-stem-cells-slow-intervertebral-disc-degeneration-through-the-NF-?B-pathway.pdf.
  7. Zhao J, Zhang Q, Wang Y, Li Y. Uterine Infusion With Bone Marrow Mesenchymal Stem Cells Improves Endometrium Thickness in a Rat Model of Thin Endometrium. Reprod Sci. 2015;22(2):181-188. doi:10.1177/1933719114537715.
  8. Fekete N, Rojewski MT, Fürst D, et al. GMP-compliant isolation and large-scale expansion of bone marrow-derived MSC. PLoS One. 2012;7(8). doi:10.1371/journal.pone.0043255.
  9. Elman JS, Li M, Wang F, Gimble JM, Parekkadan B. A comparison of adipose and bone marrow-derived mesenchymal stromal cell secreted factors in the treatment of systemic inflammation. 2014:4-11.
  10. Li C, Wu X, Tong J, et al. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res Ther. 2015;6(1):55. doi:10.1186/s13287-015-0066-5.
  11. Books J, Sign R. Safety of Intracavernous Bone Marrow-Mononuclear Cells for Postrad ... Safety of Intracavernous Bone Marrow-Mononuclear Cells for Postradical Prostatectomy Erectile Dysfunction?: An Open Dose-Escalation Pilot Study Safety of Intracavernous Bone Marrow-Mon. 2017:2015-2017.
  12. Rinker TE, Hammoudi TM, Kemp ML, Lu H, Temenoff JS. Interactions between mesenchymal stem cells, adipocytes, and osteoblasts in a 3D tri-culture model of hyperglycemic conditions in the bone marrow microenvironment. Integr Biol (Camb). 2014;6(3):324-337. doi:10.1039/c3ib40194d.
  13. Al-sayegh H, Bashir J, Goodyear S, Freeman MD. A prospective multi-site registry study of a specific protocol of autologous bone marrow concentrate for the treatment of shoulder rotator cuff tears and osteoarthritis. 2015:269-276.
  14. Rager TM, Olson JK, Zhou Y, Wang Y, Besner GE. Exosomes secreted from bone marrow-derived mesenchymal stem cells protect the intestines from experimental necrotizing enterocolitis. J Pediatr Surg. 2016;51(6):942-947. doi:10.1016/j.jpedsurg.2016.02.061.
  15. Tate-oliver K, Alexander RW. Density Platelet-Rich Plasma or Bone Marrow.
  16. Tang K, Yan J, Shen Y, et al. Tracing type 1 diabetic Tibet miniature pig ? s bone marrow mesenchymal stem cells in vitro by magnetic resonance imaging. 2014;6:123-131. doi:10.1111/1753-0407.12084.
  17. Wang X, Mamillapalli R, Mutlu L, Du H, Taylor HS. Chemoattraction of bone marrow-derived stem cells towards human endometrial stromal cells is mediated by estradiol regulated CXCL12 and CXCR4 expression. Stem Cell Res. 2015;15(1):14-22. doi:10.1016/j.scr.2015.04.004.
  18. Rambaldi A, Capelli C, Domenghini M, et al. Human platelet lysate allows expansion and clinical grade production of mesenchymal stromal cells from small samples of bone marrow aspirates or marrow filter washouts. 2007:785-791. doi:10.1038/sj.bmt.1705798.
  19. Mushtaq M, Williams AR, Suncion VY, et al. Transendocardial Mesenchymal Stem Cells and Mononuclear Bone Marrow Cells for Ischemic Cardiomyopathy The TAC-HFT Randomized Trial. 2015;16960. doi:10.1001/jama.2013.282909.
  20. 2014 Autografting of bone marrow mesenchymal stem cells alleviates streptozotocin induced diabetes in miniature pigs.pdf.
  21. Program T, Marga- P, Kingdom U. Bone Marrow Therapies for Chronic Heart Dis- ease. 2015:1-12. doi:10.1002/stem.2080.
  22. Biology C, Cell R, Eye N, Institutes N, Infor- AS. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. 2017:1273-1285. doi:10.1002/sctm.12056.
  23. Jeong Y, Kyu H, Hwa H, Chan Y. Cellular Physiology and Biochemistr y Biochemistry Direct Comparison of Human Mesenchymal Stem Cells Derived from Adipose Tissues and Bone Marrow in Mediating Neovascularization in Response to Vascular Ischemia. Cell Physiol Biochem. 2007;20:867-876.
  24. Shutian S, Shaoping N, Xingxin W, et al. GW25-e3198 The combination of transforming growth factor ?1 and 5-azacytidine improve the differentiation effects of rat Bone marrow mesenchymal stem cells into cardiomyocytes. J Am Coll Cardiol. 2014;64(16):C21. doi:10.1016/j.jacc.2014.06.105.
  25. Narita T, Suzuki K. Bone marrow-derived mesenchymal stem cells for the treatment of heart failure. Heart Fail Rev. 2014:53-68. doi:10.1007/s10741-014-9435-x.
  26. Elman JS, Li M, Wang F, Gimble JM, Parekkadan B. A comparison of adipose and bone marrow-derived mesenchymal stromal cell secreted factors in the treatment of systemic inflammation. J Inflamm (Lond). 2014;11:1. doi:10.1186/1476-9255-11-1.
  27. Dong X, Zhu F, Liu Q, et al. Transplanted bone marrow mesenchymal stem cells protects myocardium by regulating 14-3-3 protein in a rat model of diabetic cardiomyopathy. 2014;7(7):3714-3723.
  28. Huang L, Wu W, Luo F. Umbilical Cord Mesenchymal Stromal Cell With Autologous Bone Marrow Cell Transplantation in Established Type 1 Diabetes?: A Pilot Randomized Controlled Open-Label Clinical Study to Assess Safety and Impact on Insulin Secretion. 2016:1-9. doi:10.2337/dc15-0171.
  29. Sanghi V, Sethi D, Harris KL, et al. International Journal of the Cardiovascular Academy Autologous bone marrow concentrate enriched in progenitor cells ? An adjuvant in the treatment of acute myocardial infarction. IJCAC. 2016. doi:10.1016/j.ijcac.2016.04.001.
  30. Leyh M, Seitz A, Dürselen L, et al. Subchondral bone influences chondrogenic differentiation and collagen production of human bone marrow-derived mesenchymal stem cells and articular chondrocytes. 2014:1-18. doi:10.1186/s13075-014-0453-9.
  31. Surgery M, Stomatological S, Material CP, et al. T ISSUE -S PECIFIC S TEM C ELLS Adiponectin Regulates Bone Marrow Mesenchymal Stem Cell Niche Through a Unique Signal Transduction Pathway?: An Approach for Treating Bone Disease in Diabetes. 2015:240-252.
  32. Li C, Wu X, Tong J, et al. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res Ther. 2015;6(1):55. doi:10.1186/s13287-015-0066-5.
  33. Mead B, Logan A, Berry M, Leadbeater W, Scheven BA. Paracrine-Mediated Neuroprotection and Neuritogenesis of Axotomised Retinal Ganglion Cells by Human Dental Pulp Stem Cells: Comparison with Human Bone Marrow and Adipose-Derived Mesenchymal Stem Cells. PLoS One. 2014;9(10):e109305. doi:10.1371/journal.pone.0109305.
  34. Cao C, Zou J, Liu X, Shapiro A. Bone marrow mesenchymal stem cells slow intervertebral disc degeneration through the NF- k B pathway. Spine J. 2015;15(3):530-538. doi:10.1016/j.spinee.2014.11.021.
  35. Li C, Wu X, Tong J, et al. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. ??? 2015. doi:10.1186/s13287-015-0066-5.
  36. Narita T, Suzuki K. Bone marrow-derived mesenchymal stem cells for the treatment of heart failure. 2015:53-68. doi:10.1007/s10741-014-9435-x.
  37. Heldman AW, DiFede DL, Fishman JE, et al. Transendocardial Mesenchymal Stem Cells and Mononuclear Bone Marrow Cells for Ischemic Cardiomyopathy. Jama. 2014;311(1):62. doi:10.1001/jama.2013.282909.
  38. Baglio SR, Rooijers K, Koppers-Lalic D, et al. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res Ther. 2015;6(1):127. doi:10.1186/s13287-015-0116-z.
  39. Islam MN, Das SR, Emin MT, et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759-765. doi:10.1038/nm.2736.
  40. Baglio SR, Rooijers K, Koppers-Lalic D, et al. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res Ther. 2015;6(1):127. doi:10.1186/s13287-015-0116-z.
  41. Bian S, Zhang L, Duan L, et al. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J Mol Med (Berl). 2014;92(4):387-397. doi:10.1007/s00109-013-1110-5.
  42. Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure therapy mediated by the trophic activities of bone marrow mesenchymal stem cells: a noninvasive therapeutic regimen. Am J Physiol Heart Circ Physiol. 2009;296(6):H1888-H1897. doi:10.1152/ajpheart.00186.2009.
  43. Czubak PB, Bojarska-junak A, Tabarkiewicz J, Putowski LB. A Modified Method of Insulin Producing Cells ? Generation from Bone Marrow-Derived Mesenchymal Stem Cells. 2014;2014:1-7. doi:10.1155/2014/628591.
  44. Baglio SR, Rooijers K, Koppers-Lalic D, et al. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res Ther. 2015;6(1):127. doi:10.1186/s13287-015-0116-z.
  45. am Esch JS, Knoefel WT, Klein M, et al. Portal application of autologous CD133+ bone marrow cells to the liver: a novel concept to support hepatic regeneration. Stem Cells. 2005;23(4):463-470. doi:10.1634/stemcells.2004-0283.
  46. Wang X, Nie S-P, Zhen L, et al. TCTAP A-156 Retrograde Coronary Vein Delivery of Basic Fibroblast Growth Enhances Bone Marrow Mesenchymal Stem Cells Engraftment for Myocardial Repair in a Canine Infarct Model. J Am Coll Cardiol. 2014;63(12):S44. doi:10.1016/j.jacc.2014.02.189.
  47. Al-sayegh H, Bashir J, Goodyear S, Freeman MD. A prospective multi-site registry study of a specific protocol of autologous bone marrow concentrate for the treatment of shoulder rotator cuff tears and osteoarthritis. 2015:269-276.
  48. Gabr MM, Zakaria MM, Refaie AF, et al. Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells into Insulin-Producing Cells?: Evidence for Further Maturation In Vivo. 2015;2015.
  49. Pai M, Zacharoulis D, Milicevic MN, et al. Autologous infusion of expanded mobilized adult bone marrow-derived CD34+ cells into patients with alcoholic liver cirrhosis. Am J Gastroenterol. 2008;103(8):1952-1958. doi:10.1111/j.1572-0241.2008.01993.x.
  50. Kushida T, Iida H. Bone marrow cell transplantation efficiently repairs tendon and ligament injuries. Front Cell Dev Biol. 2014;2(July):1-4. doi:10.3389/fcell.2014.00027.
  51. Schulte J, Knoefel T, Klein M, et al. Portal Application of Autologous CD133 + Bone Marrow Cells to the. 2005:463-470. doi:10.1634/stemcells.2004-0283.
  52. Scott M, Ph D, Marley SB, et al. Autologous Infusion of Expanded Mobilized Adult Bone Marrow-Derived CD34 + Cells Into Patients With Alcoholic Liver Cirrhosis. 2008:1952-1958. doi:10.1111/j.1572-0241.2008.01993.x.
  53. 2015 Murine Sca1+Lin? bone marrow contains an endodermal precursor population that differentiates into hepatocytes.pdf.
  54. Prabhakar S, Marwaha N, Lal V, Sharma RR, Rajan R, Khandelwal N. Autologous bone marrow-derived stem cells in amyotrophic lateral sclerosis: A pilot study. Neurol India. 2012;60(5):465-469. doi:10.4103/0028-3886.103185.
  55. Naaldijk Y, Jäger C, Fabian C, et al. Effect of systemic transplantation of bone marrow-derived mesenchymal stem cells on neuropathology markers in APP/PS1 Alzheimer mice. Neuropathol Appl Neurobiol. 2016:1-16. doi:10.1111/nan.12319.
  56. Nicola M Di, Carlo-stella C, Magni M, et al. induced by cellular or nonspecific mitogenic stimuli Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. 2013;99(10):3838-3843. doi:10.1182/blood.V99.10.3838.
  57. Tzameret A, Sher I, Belkin M, et al. Epiretinal transplantation of human bone marrow mesenchymal stem cells rescues retinal and vision function in a rat model of retinal degeneration. Stem Cell Res. 2015;15(2):387-394. doi:10.1016/j.scr.2015.08.007.
  58. Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007;56(4):1175-1186. doi:10.1002/art.22511.
  59. Capelli C, Domenghini M, Borleri G, et al. Human platelet lysate allows expansion and clinical grade production of mesenchymal stromal cells from small samples of bone marrow aspirates or marrow filter washouts. Bone Marrow Transplant. 2007;40(8):785-791. doi:10.1038/sj.bmt.1705798.
  60. Sanghi V, Sethi D, Harris KL, et al. International Journal of the Cardiovascular Academy Autologous bone marrow concentrate enriched in progenitor cells ? An adjuvant in the treatment of acute myocardial infarction. IJCAC. 2016. doi:10.1016/j.ijcac.2016.04.001.
  61. Oe K, Kushida T, Okamoto N, et al. New strategies for anterior cruciate ligament partial rupture using bone marrow transplantation in rats. Stem Cells Dev. 2011;20(4):671-679. doi:10.1089/scd.2010.0182.
  62. Ahmed HH, Salem AM, Atta HM, et al. Updates in the pathophysiological mechanisms of Parkinson?s disease: Emerging role of bone marrow mesenchymal stem cells. World J Stem Cells. 2016;8(3):106. doi:10.4252/wjsc.v8.i3.106.
  63. Cai J, Wu Z, Xu X, et al. Umbilical Cord Mesenchymal Stromal Cell With Autologous Bone Marrow Cell Transplantation in Established Type 1 Diabetes: A Pilot Randomized Controlled Open-Label Clinical Study to Assess Safety and Impact on Insulin Secretion. Diabetes Care. 2015:dc150171. doi:10.2337/dc15-0171.
  64. Abdel Aziz MT, Wassef MAA, Ahmed HH, et al. The role of bone marrow derived-mesenchymal stem cells in attenuation of kidney function in rats with diabetic nephropathy. Diabetol Metab Syndr. 2014;6(1):34. doi:10.1186/1758-5996-6-34.
  65. Al-sayegh H, Bashir J, Goodyear S, Freeman MD. A prospective multi-site registry study of a specific protocol of autologous bone marrow concentrate for the treatment of shoulder rotator cuff tears and osteoarthritis. 2015:269-276.
  66. Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure therapy mediated by the trophic activities of bone marrow mesenchymal stem cells: a noninvasive therapeutic regimen. Am J Physiol Heart Circ Physiol. 2009;296(6):H1888-H1897. doi:10.1152/ajpheart.00186.2009.
  67. Collino F, Deregibus MC, Bruno S, et al. Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs. PLoS One. 2010;5(7). doi:10.1371/journal.pone.0011803.
  68. Sanghi V, Sethi D, Harris KL, et al. International Journal of the Cardiovascular Academy Autologous bone marrow concentrate enriched in progenitor cells ? An adjuvant in the treatment of acute myocardial infarction. IJCAC. 2016. doi:10.1016/j.ijcac.2016.04.001.
  69. Fekete N, Rojewski MT, Fürst D, et al. GMP-compliant isolation and large-scale expansion of bone marrow-derived MSC. PLoS One. 2012;7(8). doi:10.1371/journal.pone.0043255.
  70. C. L, S.A. M, M. A, S.H. V, A. F-G. Exosomes mediate the cytoprotective effects of bone Marrow-Derived Stromal Cells (MSCS) on the hypoxic lung. Am J Respir Crit Care Med. 2011;183(1 MeetingAbstracts):no pagination. http://ajrccm.atsjournals.org/cgi/reprint/183/1_MeetingAbstracts/A3764?sid=f0b58cd0-9f08-401b-bb88-e9a3268d044f%5Cnhttp://ovidsp.ovid.com/ovidweb.cgi?T=JS&PAGE=reference&D=emed10&NEWS=N&AN=70848115.
  71. Islam MN, Das SR, Emin MT, et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759-765. doi:10.1038/nm.2736.
  72. Yoon SH, Shim YS, Park YH, et al. Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage-colony stimulating factor: Phase I/II clinical trial. Stem Cells. 2007;25(8):2066-2073. doi:10.1634/stemcells.2006-0807.
  73. Hernigou P, Guissou I, Homma Y, et al. Percutaneous injection of bone marrow mesenchymal stem cells for ankle non-unions decreases complications in patients with diabetes. Int Orthop. 2015. doi:10.1007/s00264-015-2738-2.
  74. Davies OG, Smith AJ, Cooper PR, Shelton RM, Scheven BA. Cryobiology The effects of cryopreservation on cells isolated from adipose , bone marrow and dental pulp tissues q. Cryobiology. 2014;69(2):342-347. doi:10.1016/j.cryobiol.2014.08.003.
  75. C. L, S.A. M, M. A, S.H. V, A. F-G. Exosomes mediate the cytoprotective effects of bone Marrow-Derived Stromal Cells (MSCS) on the hypoxic lung. Am J Respir Crit Care Med. 2011;183(1 MeetingAbstracts):no pagination. http://ajrccm.atsjournals.org/cgi/reprint/183/1_MeetingAbstracts/A3764?sid=f0b58cd0-9f08-401b-bb88-e9a3268d044f%5Cnhttp://ovidsp.ovid.com/ovidweb.cgi?T=JS&PAGE=reference&D=emed10&NEWS=N&AN=70848115.
  76. Tang KX, Shen YF, Li BY, et al. Tracing type 1 diabetic Tibet miniature pig?s bone marrow mesenchymal stem cells in vitro by magnetic resonance imaging. J Diabetes. 2013;6:123-131. doi:10.1111/1753-0407.12084.
  77. Associates RM, Biosciences C. T RANSLATIONAL AND C LINICAL Percutaneous Injection of Autologous Bone Marrow Concentrate Cells Significantly Reduces Lumbar Discogenic Pain Through 12 Months. 2015:146-156.
  78. Kasahara Y, Matsuyama T, Taguchi A. Treatment of Autologous Bone Marrow Mononuclear Cells for Acute and Subacute Stroke Cell Therapy for Acute / Subacute Stroke. 2015:37-46. doi:10.1007/978.
  79. Qi X, Zhang J, Yuan H, et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells repair critical-sized bone defects through enhanced angiogenesis and osteogenesis in osteoporotic rats. Int J Biol Sci. 2016;12(7):836-849. doi:10.7150/ijbs.14809.
  80. Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure therapy mediated by the trophic activities of bone marrow mesenchymal stem cells: a noninvasive therapeutic regimen. Am J Physiol Heart Circ Physiol. 2009;296(6):H1888-H1897. doi:10.1152/ajpheart.00186.2009.
  81. Nicola M Di, Carlo-stella C, Magni M, et al. induced by cellular or nonspecific mitogenic stimuli Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. 2013;99(10):3838-3843. doi:10.1182/blood.V99.10.3838.
  82. Capelli C, Domenghini M, Borleri G, et al. Human platelet lysate allows expansion and clinical grade production of mesenchymal stromal cells from small samples of bone marrow aspirates or marrow filter washouts. Bone Marrow Transplant. 2007;40(8):785-791. doi:10.1038/sj.bmt.1705798.
  83. Yiou R, Hamidou L, Birebent B, et al. Safety of Intracavernous Bone Marrow-Mononuclear Cells for Postradical Prostatectomy Erectile Dysfunction: An Open Dose-Escalation Pilot Study. Eur Urol. 2016;69(6):988-991. doi:10.1016/j.eururo.2015.09.026.
  84. Neural I, Medicine R, Science M. Complete Spinal Cord Injury Treatment Using Autologous Bone Marrow Cell Transplantation and Bone Marrow Stimulation with Granulocyte Macrophage-Colony Stimulating Factor?: Phase I / II Clinical Trial. 2007:2066-2073. doi:10.1634/stemcells.2006-0807.
  85. Collino F, Deregibus MC, Bruno S, et al. Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs. PLoS One. 2010;5(7). doi:10.1371/journal.pone.0011803.
  86. Yu L, Tu Q, Han Q, et al. Adiponectin Regulates Bone Marrow Mesenchymal Stem Cell Niche Through a Unique Signal Transduction Pathway?: An Approach for Treating Bone Disease in Diabetes. Stem Cells. 2015;33:240-252. doi:10.1002/stem.1844.
  87. Lyra AC, Soares MB, da Silva LF, et al. Feasibility and safety of autologous bone marrow mononuclear cell transplantation in patients with advanced chronic liver disease. World J Gastroenterol. 2007;13(7):1067-1073. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17373741.
  88. Song F, Tang J, Geng R, et al. Comparison of the efficacy of bone marrow mononuclear cells and bone mesenchymal stem cells in the treatment of osteoarthritis in a sheep model. 2014;7(4):1415-1426.
  89. Nakamura-ishizu A, Takubo K, Kobayashi H, Suzuki-inoue K. CLEC-2 in megakaryocytes is critical for maintenance of hematopoietic stem cells in the bone marrow. 2015. doi:10.1084/jem.20150057.
  90. Leyh M, Seitz A, Dürselen L, et al. Subchondral bone influences chondrogenic differentiation and collagen production of human bone marrow-derived mesenchymal stem cells and articular chondrocytes. 2014:1-18. doi:10.1186/s13075-014-0453-9.
  91. Beane OS, Fonseca VC, Cooper LL, Koren G. Impact of Aging on the Regenerative Properties of Bone Marrow- , Muscle- , and Adipose-Derived Mesenchymal Stem / Stromal Cells. 2014:1-22. doi:10.1371/journal.pone.0115963.
  92. Nicola M Di, Carlo-stella C, Magni M, et al. induced by cellular or nonspecific mitogenic stimuli Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. 2013;99(10):3838-3843. doi:10.1182/blood.V99.10.3838.
  93. Ribeiro A, Laranjeira P, Mendes S, et al. 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(5):125. doi:10.1186/scrt336.
  94. Observer C, Alto P, Program T, Hospital M. T RANSLATIONAL AND C LINICAL Bone Marrow Therapies for Chronic Heart Disease. 2015:3212-3227. doi:10.1002/stem.2080.
  95. Beane OS, Fonseca VC, Cooper LL, Koren G. Impact of Aging on the Regenerative Properties of Bone Marrow- , Muscle- , and Adipose-Derived Mesenchymal Stem / Stromal Cells. 2014:1-22. doi:10.1371/journal.pone.0115963.
  96. Rodriguez-Menocal L, Shareef S, Salgado M, Shabbir A, Van Badiavas E. Role of whole bone marrow, whole bone marrow cultured cells, and mesenchymal stem cells in chronic wound healing. Stem Cell Res Ther. 2015;6(1):1-11. doi:10.1186/s13287-015-0001-9.
  97. Islam MN, Das SR, Emin MT, et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759-765. doi:10.1038/nm.2736.
  98. Venkataramana NK, Kumar SK V, Balaraju S, et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson?s disease. Transl Res. 2010;155(2):62-70. doi:10.1016/j.trsl.2009.07.006.
  99. Czubak P, Bojarska-Junak A, Tabarkiewicz J, Putowski L. A modified method of insulin producing cells? generation from bone marrow-derived mesenchymal stem cells. J Diabetes Res. 2014;2014:628591. doi:10.1155/2014/628591.
  100. Shutian S, Shaoping N, Xingxin W, et al. GW25-e3198 The combination of transforming growth factor ?1 and 5-azacytidine improve the differentiation effects of rat Bone marrow mesenchymal stem cells into cardiomyocytes. J Am Coll Cardiol. 2014;64(16):C21. doi:10.1016/j.jacc.2014.06.105.
  101. Yang J, Kaur K, Ong LL, Eisenberg CA, Eisenberg LM. Inhibition of G9a Histone Methyltransferase Converts Bone Marrow Mesenchymal Stem Cells to Cardiac Competent Progenitors. Stem Cells Int. 2015;2015:1-12. doi:10.1155/2015/270428.
  102. Biology C, Cell R, Eye N, Institutes N, Infor- AS. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. 2017. doi:10.1002/sctm.12056.
  103. Chen J, Venkat P, Chopp M. Bone Marrow Mesenchymal Stromal Cell Transplantation?: A Neurorestorative Therapy for Stroke. 2015:47-69. doi:10.1007/978.
  104. Kondo M, Kamiya H, Himeno T, et al. Therapeutic efficacy of bone marrow-derived mononuclear cells in diabetic polyneuropathy is impaired with aging or diabetes. J Diabetes Investig. 2015;6(2):140-149. doi:10.1111/jdi.12272.
  105. Nakamura-ishizu A, Takubo K, Kobayashi H, Suzuki-inoue K. CLEC-2 in megakaryocytes is critical for maintenance of hematopoietic stem cells in the bone marrow. 2015. doi:10.1084/jem.20150057.
  106. Gabr MM, Zakaria MM, Refaie AF, et al. Generation of insulin-producing cells from human bone marrow-derived mesenchymal stem cells: comparison of three differentiation protocols. Biomed Res Int. 2014;2014:832736. doi:10.1155/2014/832736.
  107. Tzameret A, Sher I, Belkin M, et al. Epiretinal transplantation of human bone marrow mesenchymal stem cells rescues retinal and vision function in a rat model of retinal degeneration ?. Stem Cell Res. 2015;15(2):387-394. doi:10.1016/j.scr.2015.08.007.
  108. C. L, S.A. M, M. A, S.H. V, A. F-G. Exosomes mediate the cytoprotective effects of bone Marrow-Derived Stromal Cells (MSCS) on the hypoxic lung. Am J Respir Crit Care Med. 2011;183(1 MeetingAbstracts):no pagination. http://ajrccm.atsjournals.org/cgi/reprint/183/1_MeetingAbstracts/A3764?sid=f0b58cd0-9f08-401b-bb88-e9a3268d044f%5Cnhttp://ovidsp.ovid.com/ovidweb.cgi?T=JS&PAGE=reference&D=emed10&NEWS=N&AN=70848115.
  109. Sun J, Wei ZZ, Gu X, et al. Intranasal delivery of hypoxia-preconditioned bone marrow-derived mesenchymal stem cells enhanced regenerative effects after intracerebral hemorrhagic stroke in mice. Exp Neurol. 2015. doi:10.1016/j.expneurol.2015.03.011.
  110. Talaat M, Aziz A, Abdel M, et al. The role of bone marrow derived-mesenchymal stem cells in attenuation of kidney function in rats with diabetic nephropathy. 2014:1-10.
  111. Song F, Tang J, Geng R, et al. Comparison of the efficacy of bone marrow mononuclear cells and bone mesenchymal stem cells in the treatment of osteoarthritis in a sheep model. 2014;7(4):1415-1426.
  112. Kallis YN, Alison MR, Forbes SJ. Bone marrow stem cells and liver disease. Gut. 2007;56(5):716-724. doi:10.1136/gut.2006.098442.
  113. Kondo M, Kamiya H, Himeno T, et al. Therapeutic ef fi cacy of bone marrow-derived mononuclear cells in diabetic polyneuropathy is impaired with aging or diabetes. 2015;6(2). doi:10.1111/jdi.12272.
  114. Czubak P, Bojarska-Junak A, Tabarkiewicz J, Putowski L. A modified method of insulin producing cells? generation from bone marrow-derived mesenchymal stem cells. J Diabetes Res. 2014;2014:628591. doi:10.1155/2014/628591.
  115. Beane OS, Fonseca VC, Cooper LL, Koren G. Impact of Aging on the Regenerative Properties of Bone Marrow- , Muscle- , and Adipose-Derived Mesenchymal Stem / Stromal Cells. 2014:1-22. doi:10.1371/journal.pone.0115963.
  116. Song F, Tang J, Geng R, et al. Comparison of the efficacy of bone marrow mononuclear cells and bone mesenchymal stem cells in the treatment of osteoarthritis in a sheep model. 2014;7(4):1415-1426.
  117. Davies OG, Smith AJ, Cooper PR, Shelton RM, Scheven BA. Cryobiology The effects of cryopreservation on cells isolated from adipose , bone marrow and dental pulp tissues q. Cryobiology. 2014;69(2):342-347. doi:10.1016/j.cryobiol.2014.08.003.
  118. Sanz-Ruiz R, Garcia AN, Elizaga J, Fdez-Aviles F. TCT-150 First-in-man Experience with Transendocardial Injections of Bone Marrow-Derived Mesenchymal Stem Cells in Idiopathic Dilated Cardiomyopathy. The MYOCYTE trial. J Am Coll Cardiol. 2014;64(11):B45. doi:10.1016/j.jacc.2014.07.186.
  119. Totey SM. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinsons disease. Transl Res. 2010;155(2):62-70. doi:10.1016/j.trsl.2009.07.006.
  120. Biology C, Cell R, Eye N, Institutes N. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. 2017:1273-1285.
  121. Biology C, Cell R, Eye N, Institutes N. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. 2017.
  122. Naaldijk Y, J C. Effect of systemic transplantation of bone marrow-derived mesenchymal stem cells on neuropathology markers in APP / PS1 Alzheimer mice. 2016:1-16. doi:10.1111/nan.12319.
  123. Development CD. C ELL -B ASED D RUG D EVELOPMENT , S CREENING , AND T OXICOLOGY Phase I Trial of Repeated Intrathecal Autologous Bone Marrow-Derived Mesenchymal Stromal Cells in Amyotrophic Lateral Sclerosis. 2015:590-597.
  124. Prabhakar S, Marwaha N, Lal V, Sharma RR, Rajan R, Khandelwal N. amyotrophic lateral sclerosis?: A pilot study Autologous bone marrow-derived stem cells in amyotrophic lateral sclerosis?: A pilot study. 2016;(September 2012). doi:10.4103/0028-3886.103185.

References and Literature - Mesenchymal Stem Cells 'MSCs'

  1. Gu W, Zhang F, Xue Q, Ma Z, Lu P, Yu B. Transplantation of bone marrow mesenchymal stem cells reduces lesion volume and induces axonal regrowth of injured spinal cord. Neuropathology. 2010; 30: 205-217.
  2. Wilkins A, Kemp K, Ginty M, Hares K, Mallam E, Scolding N. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Res. 2009; 3: 6370
  3. Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin. 2013; 34: 747-754.
  4. Wang S, Qu X, Zhao RC. Clinical applications of mesenchymal stem cells. J Hematol Oncol. 2014; 5: 19.
  5. Farini, Andrea, et al. "Clinical applications of mesenchymal stem cells in chronic diseases." Stem cells international 2014 (2014).
  6. Volarevic, Vladislav, et al. "Concise review: therapeutic potential of mesenchymal stem cells for the treatment of acute liver failure and cirrhosis." Stem Cells 32.11 (2014): 2818-2823.
  7. Ikebe, Chiho, and Ken Suzuki. "Mesenchymal stem cells for regenerative therapy: optimization of cell preparation protocols." BioMed research international 2014 (2014).
  8. Sharma, Ratti Ram, et al. "Mesenchymal stem or stromal cells: a review of clinical applications and manufacturing practices." Transfusion 54.5 (2014): 1418-1437.
  9. Jo, Chris Hyunchul, et al. "Intra‐articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof‐of‐concept clinical trial." Stem cells 32.5 (2014): 1254-1266.
  10. Squillaro, Tiziana, Gianfranco Peluso, and Umberto Galderisi. "Clinical trials with mesenchymal stem cells: an update." Cell transplantation 25.5 (2016): 829-848.
  11. Orozco, Lluis, et al. "Treatment of knee osteoarthritis with autologous mesenchymal stem cells: two-year follow-up results." Transplantation 97.11 (2014): e66-e68.
  12. Filardo, Giuseppe, et al. "Mesenchymal stem cells for the treatment of cartilage lesions: from preclinical findings to clinical application in orthopaedics." Knee surgery, sports traumatology, arthroscopy 21.8 (2013): 1717-1729.
  13. Jo, Chris Hyunchul, et al. "Intra‐articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof‐of‐concept clinical trial." Stem cells 32.5 (2014): 1254-1266.
  14. Vangsness, C. Thomas, et al. "Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy." J Bone Joint Surg Am 96.2 (2014): 90-98.

References and Literature - Platelet-Rich Plasma 'PRP' (Click for more)

  1. Laver, Lior, et al. "PRP for Degenerative Cartilage Disease A Systematic Review of Clinical Studies." Cartilage (2016): 1947603516670709.
  2. Fabbrocini, Gabriella, et al. "PRP for Lip and Eye Rejuvenation." Nonsurgical Lip and Eye Rejuvenation Techniques. Springer International Publishing, 2016. 77-83.
  3. Meheux, Carlos J., et al. "Efficacy of intra-articular platelet-rich plasma injections in knee osteoarthritis: a systematic review." Arthroscopy: The Journal of Arthroscopic & Related Surgery 32.3 (2016): 495-505.
  4. Malavolta, Eduardo A., et al. "Comments on: Evaluation of platelet-rich plasma and fibrin matrix to assist in healing and repair of rotator cuff injuries: a systematic review and meta-analysis." Clinical rehabilitation 30.7 (2016): 726-727.
  5. Zhou, Yiqin, and James HC Wang. "PRP Treatment Efficacy for Tendinopathy: A Review of Basic Science Studies." BioMed Research International 2016 (2016).
  6. Sampson S, Gerhardt M, Mandelbaum B. Platelet rich plasma injection grafts for musculoskeletal injuries: a review. Curr Rev Musculoskelet Med. 2008;1(3-4):165-174. doi:10.1007/s12178-008-9032-5.
  7. Amini F, Ramasamy TS. Efficacy of platelet rich plasma ( PRP ) on skin rejuvenation : A systematic review. 2015;18(3):119-127.
  8. Sanchez AR, Sheridan PJ, Kupp LI. Is platelet rich plasma the perfect enhancement factor? A current review. Int J Oral Maxillo- fac Surg. 2003;18:93?103
  9. Sampson S, Gerhardt M, Mandelbaum B. Platelet rich plasma injection grafts for musculoskeletal injuries: a review. Curr Rev Musculoskelet Med. 2008;1(3-4):165-174. doi:10.1007/s12178-008-9032-5.

References and Literature - Stem Cell-based Therapies and Neurodegenerative Diseases (Click for more)

  1. Tanna, Tanmay, and Vatsal Sachan. "Mesenchymal stem cells: potential in treatment of neurodegenerative diseases." Current stem cell research & therapy 9.6 (2014): 513-521.
  2. Li, Matthew D., Harold Atkins, and Tania Bubela. "The global landscape of stem cell clinical trials." Regenerative medicine 9.1 (2014): 27-39.
  3. Drago, Denise, et al. "The stem cell secretome and its role in brain repair." Biochimie 95.12 (2013): 2271-2285.
  4. Silva, Andreia M., et al. "Extracellular vesicles: immunomodulatory messengers in the context of tissue repair/regeneration." European Journal of Pharmaceutical Sciences 98 (2017): 86-95.]
  5. Yang, Yongxiang, et al. "MSCs-Derived Exosomes and Neuroinflammation, Neurogenesis and Therapy of Traumatic Brain Injury." Frontiers in Cellular Neuroscience 11 (2017).
  6. Salgado, Antonio J., et al. "Mesenchymal stem cells secretome as a modulator of the neurogenic niche: basic insights and therapeutic opportunities." Frontiers in cellular neuroscience 9 (2015).
  7. Kim, Hyun Ok, Seong-Mi Choi, and Han-Soo Kim. "Mesenchymal stem cell-derived secretome and microvesicles as a cell-free therapeutics for neurodegenerative disorders." Tissue Engineering and Regenerative Medicine 10.3 (2013): 93-101.
  8. Teixeira, Fábio G., et al. "Mesenchymal stem cells secretome: a new paradigm for central nervous system regeneration?." Cellular and Molecular Life Sciences 70.20 (2013): 3871-3882.

References and Literature - Stem Cell-based Therapies and Knee Injuries (Click for more)

  1. Kahn, Timothy L., and Ran Schwarzkopf. "Do Total Knee Arthroplasty Patients Have a Higher Activity Level Compared to Patients With Osteoarthritis?." Geriatric Orthopaedic Surgery & Rehabilitation 7.3 (2016): 142-147.
  2. Skou, Søren T., et al. "A randomized, controlled trial of total knee replacement." New England Journal of Medicine 373.17 (2015): 1597-1606.
  3. Metsna, Vahur, Sigrid Vorobjov, and Aare Märtson. "Prevalence of anterior knee pain among patients following total knee arthroplasty with nonreplaced patella: a retrospective study of 1778 knees." Medicina 50.2 (2014): 82-86.
  4. Riddle, Daniel L., William A. Jiranek, and Curtis W. Hayes. "Use of a validated algorithm to judge the appropriateness of total knee arthroplasty in the United States: a multicenter longitudinal cohort study." Arthritis & Rheumatology 66.8 (2014): 2134-2143.
  5. Wylde, Vikki, et al. "Persistent pain after joint replacement: prevalence, sensory qualities, and postoperative determinants." PAIN® 152.3 (2011): 566-572.
  6. Fuzier, Régis, et al. "Analgesic drug consumption increases after knee arthroplasty: a pharmacoepidemiological study investigating postoperative pain." PAIN® 155.7 (2014): 1339-1345. International Osteoporosis Foundation
  7. Lalmohamed, Arief, et al. "Changes in mortality patterns following total hip or knee arthroplasty over the past two decades: a nationwide cohort study." Arthritis & Rheumatology 66.2 (2014): 311-318.
  8. Lalmohamed, Arief, et al. "Timing of acute myocardial infarction in patients undergoing total hip or knee replacement: a nationwide cohort study." Archives of internal medicine 172.16 (2012): 1229-1235.
  9. Singh, Jasvinder A., et al. "Smoking as a risk factor for short‐term outcomes following primary total hip and total knee replacement in veterans." Arthritis care & research 63.10 (2011): 1365-1374.
  10. Houdek MT. Mesenchymal stem cell therapy for osteoarthritis : current perspectives. 2015:117-124.
  11. Tang J, Cui W, Song F, Zhai C. Effects of mesenchymal stem cells on interleukin-1 β -treated chondrocytes and cartilage in a rat osteoarthritic model. 2015:1753-1760. doi:10.3892/mmr.2015.3645.
  12. Mazor M, Lespessailles E, Coursier R. Mesenchymal stem-cell potential in cartilage repair : an update Osteoarthritis. 2014;18(12):2340-2350. doi:10.1111/jcmm.12378.
  13. Song F, Tang J, Geng R, et al. Comparison of the efficacy of bone marrow mononuclear cells and bone mesenchymal stem cells in the treatment of osteoarthritis in a sheep model. 2014;7(4):1415-1426.
  14. Ham O, Lee CY, Kim R, et al. Therapeutic Potential of Differentiated Mesenchymal Stem Cells for Treatment of Osteoarthritis. 2015;(April):14961-14978. doi:10.3390/ijms160714961.
  15. Kristjánsson B, Honsawek S. Current Perspectives in Mesenchymal Stem Cell Therapies for Osteoarthritis. 2014;2014. doi:10.1155/2014/194318.
  16. Centeno, Christopher, et al. "Efficacy of autologous bone marrow concentrate for knee osteoarthritis with and without adipose graft." BioMed research international 2014 (2014).
  17. Toh WS, Foldager CB, Pei M. Advances in Mesenchymal Stem Cell-based Strategies for Cartilage Repair and Regeneration. Stem Cell Rev Rep. 2014;10:686-696. doi:10.1007/s12015-014-9526-z.
  18. Centeno, Christopher J., et al. "Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells, platelet lysate and dexamethasone." Annals of Transplantation 9 (2008): 246-251.
  19. Orozco, Lluis, et al. "Treatment of knee osteoarthritis with autologous mesenchymal stem cells: two-year follow-up results." Transplantation 97.11 (2014): e66-e68.
  20. Filardo, Giuseppe, et al. "Mesenchymal stem cells for the treatment of cartilage lesions: from preclinical findings to clinical application in orthopaedics." Knee surgery, sports traumatology, arthroscopy 21.8 (2013): 1717-1729.
  21. Jo, Chris Hyunchul, et al. "Intra‐articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof‐of‐concept clinical trial." Stem cells 32.5 (2014): 1254-1266.
  22. Vangsness, C. Thomas, et al. "Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy." J Bone Joint Surg Am 96.2 (2014): 90-98.

References and Literature - Stem Cell-based Therapies for Amythrophic Aateral Sclerosis-ALS

  1. Wijesekera, Lokesh C., and P. Nigel Leigh. "Amyotrophic lateral sclerosis." Orphanet journal of rare diseases 4.1 (2009): 3.
  2. Ferraiuolo, Laura, et al. "Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis." Nature Reviews Neurology 7.11 (2011): 616-630.
  3. Mazzini, Letizia, et al. "Stem cell therapy in amyotrophic lateral sclerosis: a methodological approach in humans." Amyotrophic Lateral Sclerosis and Other Motor Neuron Disorders 4.3 (2003): 158-161.
  4. Mazzini, L., et al. "Mesenchymal stem cell transplantation in amyotrophic lateral sclerosis: A Phase I clinical trial." Experimental neurology 223.1 (2010): 229-237.
  5. Papadeas, Sophia T., and Nicholas J. Maragakis. "Advances in stem cell research for Amyotrophic Lateral Sclerosis." Current opinion in Biotechnology 20.5 (2009): 545-551.
  6. Janson, C. G., et al. "Human intrathecal transplantation of peripheral blood stem cells in amyotrophic lateral sclerosis." Journal of hematotherapy & stem cell research 10.6 (2001): 913-915.
  7. Thomsen, Gretchen M., et al. "The past, present and future of stem cell clinical trials for ALS." Experimental neurology 262 (2014): 127-137
  8. Staff, Nathan P., et al. "Safety of intrathecal autologous adipose-derived mesenchymal stromal cells in patients with ALS." Neurology 87.21 (2016): 2230-2234.
  9. Oh, Ki-Wook, et al. "Phase I Trial of Repeated Intrathecal Autologous Bone Marrow Derived Mesenchymal Stromal Cells in Amyotrophic Lateral Sclerosis." Stem cells translational medicine 4.6 (2015): 590-597.
  10. Petrou, Panayiota, et al. "Safety and clinical effects of mesenchymal stem cells secreting neurotrophic factor transplantation in patients with amyotrophic lateral sclerosis: results of phase 1/2 and 2a clinical trials." JAMA neurology 73.3 (2016): 337-344.
  11. Farinazzo, Alessia, et al. "Murine adipose-derived mesenchymal stromal cell vesicles: in vitro clues for neuroprotective and neuroregenerative approaches." Cytotherapy 17.5 (2015): 571-578.
  12. Bonafede, Roberta, et al. "Exosome derived from murine adipose-derived stromal cells: Neuroprotective effect on in vitro model of amyotrophic lateral sclerosis." Experimental cell research 340.1 (2016): 150-158.
  13. Boruczkowski, D., et al. "Mesenchymal Stem Cells As A Therapeutic Option For Patients With ALS." Gen Med (Los Angel) 4.235 (2016): 2.

References and Literature - Stem Cell-based Therapies and Anti-Aging

  1. Dong, Xiao, Brandon Milholland, and Jan Vijg. “Evidence for a limit to human lifespan.” Nature 538.7624 (2016): 257.
  2. https://health.usnews.com/health-news/patient-advice/articles/2015/01/30/hospital-labs-behind-the-scenes
  3. Farr, J. Michael, and Laurence Shatkin. Best jobs for the 21st century. Indianapolis, IN: Jist Works, 2009. ISBN 978-1-56370-961-6
  4. Weidner, Carola Ingrid, et al. “Aging of blood can be tracked by DNA methylation changes at just three CpG sites.” Genome biology 15.2 (2014): R24.
  5. Eipel, Monika, et al. “Epigenetic age predictions based on buccal swabs are more precise in combination with cell type-specific DNA methylation signature
  6. Hayflick, Leonard. “The limited in vitro lifetime of human diploid cell strains.” Experimental cell research 37.3 (1965): 614-636.
  7. 10 Butler, Merlin G., et al. “Comparison of chromosome telomere integrity in multiple tissues from subjects at different ages.” Cancer genetics and cytogenetics 105.2 (1998): 138-144.
  8. 11 Lindsey, Janet, et al. “In vivo loss of telomeric repeats with age in humans.” Mutation Research/DNAging 256.1 (1991): 45-48.
  9. 12 Aubert, G. “Lansdorp pM. Telomeres and aging. 37.” Physiol Rev 88 (2008): 557-79.
  10. 13 Daniali, Lily, et al. “Telomeres shorten at equivalent rates in somatic tissues of adults.” Nature communications 4 (2013): 1597.
  11. 14 Castelo-Branco, Camil, and Iris Soveral. “The immune system and aging: a review.” Gynecological Endocrinology 30.1 (2014): 16-22.
  12. 15 Desai, Anjali, Annabelle Grolleau-Julius, and Raymond Yung. “Leukocyte function in the aging immune system.” Journal of leukocyte biology 87.6 (2010): 1001-1009.
  13. 16 Frasca, Daniela, and Bonnie B. Blomberg. “Aging, cytomegalovirus (CMV) and influenza vaccine responses.” Human vaccines & immunotherapeutics 12.3 (2016): 682-690.
  14. 17 Mazzola, Paolo, et al. “Aging, cancer, and cancer vaccines.” Immunity & Ageing 9.1 (2012): 4.
  15. 18 Baylis, Daniel, et al. “Understanding how we age: insights into inflammaging.” Longevity & healthspan 2.1 (2013): 8.
  16. Werner, Christian M., et al. “Differential effects of endurance, interval, and resistance training on telomerase activity and telomere length in a randomized, controlled study.” European heart journal 40.1 (2018): 34-46.
  17. Dietary Supplement Labels: Key Elements Office of Inspector General, Depart of Health and Human Services. 2003.
  18. National Institutes of Health. “Dietary supplements: Background information.” Office of Dietary Supplements Fact Sheet. Washington, DC: National Institutes of Health, US Department of Health and Human Services (2011).
  19. LIEBERMAN, Shari, and Nancy BRUNING. “The Real Vitamin & Mineral Book-Going beyond the RDA for Optimum Health; Avery Publ.” Group (1990): 142.
  20. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). “Scientific Opinion in relation to the authorisation procedure for health claims on calcium and vitamin D and the reduction of the risk of osteoporotic fractures by reducing bone loss pursuant to Article 14 of Regulation (EC) No 1924/2006.” EFSA Journal 8.5 (2010): 1609.
  21. Food and Drug Administration, HHS. “Food labeling: health claims; calcium and osteoporosis, and calcium, vitamin D, and osteoporosis. Final rule.” Federal register 73.189 (2008): 56477.
  22. US Food and Drug Administration. “Qualified health claims: letter of enforcement discretion–nuts and coronary heart disease.” Rockville, MD: US Food and Drug Administration (2003): 1-4.
  23. Thomas, D. Travis, Kelly Anne Erdman, and Louise M. Burke. “Position of the Academy of Nutrition and Dietetics, Dietitians of Canada, and the American College of Sports Medicine: nutrition and athletic performance.” Journal of the Academy of Nutrition and Dietetics 116.3 (2016): 501-528.
  24. Komar, B., L. Schwingshackl, and Georg Hoffmann. “Effects of leucine-rich protein supplements on anthropometric parameter and muscle strength in the elderly: a systematic review and meta-analysis.” The journal of nutrition, health & aging 19.4 (2015): 437-446.
  25. Dong, Jia-Yi, et al. “Effect of oral L-arginine supplementation on blood pressure: a meta-analysis of randomized, double-blind, placebo-controlled trials.” American heart journal 162.6 (2011): 959-965.
  26. Omega−3 Fatty Acids and Health: Fact Sheet for Health Professionals. US National Institutes of Health, Office of Dietary Supplements. 2 November 2016. Retrieved 5 April 2017
  27. European Food Safety Authority (EFSA). “Dietary Reference Values for nutrients Summary report.” EFSA Supporting Publications 14.12 (2017): e15121E.
  28. Siscovick, David S., et al. “Omega-3 polyunsaturated fatty acid (fish oil) supplementation and the prevention of clinical cardiovascular disease: a science advisory from the American Heart Association.” Circulation 135.15 (2017): e867-e884.
  29. Adapted from: Life Extension, 3600 West Commercial Boulevard, Fort Lauderdale, FL 33309
  30. Barzilai, Nir, et al. “Metformin as a tool to target aging.” Cell metabolism 23.6 (2016): 1060-1065.
  31. Available at: https://www.drugs.com/dosage/metformin.html.
  32. Martin-Montalvo, Alejandro, et al. “Metformin improves healthspan and lifespan in mice.” Nature communications 4 (2013): 2192.
  33. Bannister, CA 1., et al. “Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched, non-diabetic controls.” Diabetes, Obesity and Metabolism 16.11 (2014): 1165-1173.
  34. De Oliveira Figueiredo, Rejane Augusta, et al. “Diabetes mellitus, metformin and head and neck cancer.” Oral oncology 61 (2016): 47-54.
  35. Tseng, Chin-Hsiao. “Metformin reduces gastric cancer risk in patients with type 2 diabetes mellitus.” Aging (Albany NY) 8.8 (2016): 1636.
  36. Coyle, C., et al. “Metformin as an adjuvant treatment for cancer: a systematic review and meta-analysis.” Annals of Oncology 27.12 (2016): 2184-2195.
  37. Anisimov, Vladimir N. “Metformin for cancer and aging prevention: is it a time to make the long story short?.” Oncotarget 6.37 (2015): 39398.
  38. Dong, Yunzhou, et al. “Activation of AMP-activated protein kinase inhibits oxidized LDL-triggered endoplasmic reticulum stress in vivo.” Diabetes 59.6 (2010): 1386-1396.
  39. Vasamsetti, Sathish Babu, et al. “Metformin inhibits monocyte-to-macrophage differentiation via AMPK-mediated inhibition of STAT3 activation: potential role in atherosclerosis.” Diabetes 64.6 (2015): 2028-2041.
  40. Wan, Xinyi, et al. “5’-AMP–Activated Protein Kinase–Activating Transcription Factor 1 Cascade Modulates Human Monocyte–Derived Macrophages to Atheroprotective Functions in Response to Heme or Metformin.” Arteriosclerosis, thrombosis, and vascular biology 33.11 (2013): 2470-2480.
  41. Abualsuod, Amjad, et al. “The effect of metformin use on left ventricular ejection fraction and mortality post-myocardial infarction.” Cardiovascular drugs and therapy 29.3 (2015): 265-275.
  42. 52 Anabtawi, Abeer, and John M. Miles. “Metformin: nonglycemic effects and potential novel indications.” Endocrine Practice 22.8 (2016): 999-1007.
  43. Wang, Man, et al. “Metformin for treatment of antipsychotic-induced weight gain: a randomized, placebo-controlled study.” Schizophrenia research 138.1 (2012): 54-57.
  44. Luchsinger, José A., et al. “Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial.” Journal of Alzheimer’s Disease 51.2 (2016): 501-514.
  45. Pérez-Revuelta, B. I., et al. “Metformin lowers Ser-129 phosphorylated α-synuclein levels via mTOR-dependent protein phosphatase 2A activation.” Cell death & disease 5.5 (2014): e1209.
  46. Bayliss, Jacqueline A., et al. “Metformin prevents nigrostriatal dopamine degeneration independent of AMPK activation in dopamine neurons.” PLoS One 11.7 (2016): e0159381.
  47. Johnson, Simon C., Peter S. Rabinovitch, and Matt Kaeberlein. “mTOR is a key modulator of ageing and age-related disease.” Nature 493.7432 (2013): 338.
  48. Miklya, I. “The significance of selegiline/(−)-deprenyl after 50 years in research and therapy (1965–2015).” Molecular psychiatry 21.11 (2016): 1499.
  49. Stuenkel, Cynthia A., et al. “Treatment of symptoms of the menopause: an endocrine society clinical practice guideline.” The Journal of Clinical Endocrinology & Metabolism 100.11 (2015): 3975-4011.
  50. Bhasin, Shalender, et al. “Testosterone therapy in men with hypogonadism: an Endocrine Society clinical practice guideline.” The Journal of Clinical Endocrinology & Metabolism 103.5 (2018): 1715-1744.
  51. Chen, Shaoliang, et al. “Intracoronary transplantation of autologous bone marrow mesenchymal stem cells for ischemic cardiomyopathy due to isolated chronic occluded left anterior descending artery.” The Journal of invasive cardiology 18.11 (2006): 552-556.
  52. Bang, Oh Young, et al. “Autologous mesenchymal stem cell transplantation in stroke patients.” Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society 57.6 (2005): 874-882.
  53. Pal, Rakhi, et al. “Ex vivo-expanded autologous bone marrow-derived mesenchymal stromal cells in human spinal cord injury/paraplegia: a pilot clinical study.” Cytotherapy 11.7 (2009): 897-911.
  54. Venkataramana, Neelam K., et al. “Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson’s disease.” Translational Research 155.2 (2010): 62-70.
  55. Harris, Violaine K., et al. “Phase I trial of intrathecal mesenchymal stem cell-derived neural progenitors in progressive multiple sclerosis.” EBioMedicine 29 (2018): 23-30.
  56. Sun, Lingyun, et al. “Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans.” Stem cells 27.6 (2009): 1421-1432.
  57. Dash, Nihar Ranjan, et al. “Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow-derived mesenchymal stem cells.” Rejuvenation research 12.5 (2009): 359-366.
  58. Wakitani, Shigeyuki, et al. “Safety of autologous bone marrow derived mesenchymal stem cell transplantation for cartilage repair in 41 patients with 45 joints followed for up to 11 years and 5 months.” Journal of Tissue Engineering and Regenerative Medicine 5.2 (2011): 146-150.
  59. Khera, Mohit, Maarten Albersen, and John P. Mulhall. “Mesenchymal stem cell therapy for the treatment of erectile dysfunction.” The journal of sexual medicine 12.5 (2015): 1105-1106.
  60. Kim, Won-Serk, et al. “Antiwrinkle effect of adipose-derived stem cell: activation of dermal fibroblast by secretory factors.” Journal of dermatological science 53.2 (2009): 96-102.
  61. Brüünsgaard, Helle, and Bente Klarlund Pedersen. “Age-related inflammatory cytokines and disease.” Immunology and Allergy Clinics 23.1 (2003): 15-39.
  62. Ferrucci, Luigi, et al. “Serum IL-6 level and the development of disability in older persons.” Journal of the American Geriatrics Society 47.6 (1999): 639-646.
  63. Visser, Marjolein, et al. “Relationship of interleukin-6 and tumor necrosis factor-α with muscle mass and muscle strength in elderly men and women: the Health ABC Study.” The Journals of Gerontology Series A: Biological Sciences and Medical Sciences 57.5 (2002): M326-M332.
  64. Kanapuru, Bindu, and William B. Ershler. “Inflammation, coagulation, and the pathway to frailty.” The American journal of medicine 122.7 (2009): 605-613.
  65. Walston, Jeremy D., et al. “Inflammation and stress-related candidate genes, plasma interleukin-6 levels, and longevity in older adults.” Experimental gerontology 44.5 (2009): 350-355.
  66. Gonzalez, Rafael, Dave Woynarowski, and Luis Geffner. “Stem cells targeting inflammation as potential anti-aging strategies and therapies.” Cell & Tissue Transplantation & Therapy 2015.7 (2015): 1-8.
  67. Fontaine, Magali J., et al. “Unraveling the mesenchymal stromal cells’ paracrine immunomodulatory effects.” Transfusion Medicine Reviews 30.1 (2016): 37-43.
  68. Chen, Wancheng, et al. “Immunomodulatory effects of mesenchymal stromal cells-derived exosome.” Immunologic research 64.4 (2016): 831-840.
  69. Kota, Daniel J., et al. “TSG-6 produced by hMSCs delays the onset of autoimmune diabetes by suppressing Th1 development and enhancing tolerogenicity.” Diabetes 62.6 (2013): 2048-2058.
  70. Shigemoto-Kuroda, Taeko, et al. “MSC-derived extracellular vesicles attenuate immune responses in two autoimmune murine models: type 1 diabetes and uveoretinitis.” Stem cell reports 8.5 (2017): 1214-1225.
  71. Freund, Adam, et al. “Inflammatory networks during cellular senescence: causes and consequences.” Trends in molecular medicine 16.5 (2010): 238-246.
  72. Coppé, Jean-Philippe, et al. “The senescence-associated secretory phenotype: the dark side of tumor suppression.” Annual Review of Pathological Mechanical Disease 5 (2010): 99-118.
  73. Roos, Carolyn M., et al. “Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice.” Aging cell 15.5 (2016): 973-977.
  74. Childs, Bennett G., et al. “Senescent intimal foam cells are deleterious at all stages of atherosclerosis.” Science 354.6311 (2016): 472-477.
  75. Xu, Ming, et al. “Transplanted senescent cells induce an osteoarthritis-like condition in mice.” The Journals of Gerontology: Series A 72.6 (2017): 780-785.
  76. Baker, Darren J., et al. “Clearance of p16 Ink4a-positive senescent cells delays ageing-associated disorders.” Nature 479.7372 (2011): 232.
  77. Tchkonia, Tamara, et al. “Cellular senescence and the senescent secretory phenotype: therapeutic opportunities.” The Journal of clinical investigation 123.3 (2013): 966-972.
  78. Zhu, Yi, et al. “The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs.” Aging cell 14.4 (2015): 644-658.
  79. Soebadi, M. Ayodhia, et al. “Advances in stem cell research for the treatment of male sexual dysfunctions.” Current opinion in urology 26.2 (2016): 129-139.
  80. Ozsvari, Bela, et al. “Azithromycin and Roxithromycin define a new family of “senolytic” drugs that target senescent human fibroblasts.” Aging (Albany NY) 10.11 (2018): 3294.
  81. Patil, S. P., et al. “Neuroprotective effect of metformin in MPTP-induced Parkinson’s disease in mice.” Neuroscience 277 (2014): 747-754.
  82. Park, Byung-Soon, and Won-Serk Kim. "Adipose-Derived Stem Cells and Their Secretory Factors for Skin Aging and Hair Loss." Textbook of Aging Skin (2017): 205-224.
  83. Xu, Dan, and Hidetoshi Tahara. "The role of exosomes and microRNAs in senescence and aging." Advanced drug delivery reviews 65.3 (2013): 368-375.
  84. Prattichizzo, Francesco, et al. "Exosome-based immunomodulation during aging: a nano-perspective on inflamm-aging." Mechanisms of Ageing and Development (2017).
  85. Basu, Joydeep, and John W. Ludlow. "Exosomes for repair, regeneration and rejuvenation." Expert opinion on biological therapy 16.4 (2016): 489-506.
  86. Park, Byung-Soon, and Won-Serk Kim. "Adipose-derived stem cells and their secretory factors for skin aging." Textbook of Aging Skin. Springer Berlin Heidelberg, 2010. 201-212.
  87. Soto-Gamez, A., & Demaria, M. (2017). Therapeutic interventions for aging: the case of cellular senescence. Drug Discovery Today.
  88. Moiseeva, O. et al. (2013) Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kB activation. Aging Cell 12, 489–498.
  89. Martin-Montalvo, A. et al. (2013) Metformin improves healthspan and lifespan in mice. Nat. Commun. 4, 2192.
  90. Pitozzi, V. et al. (2013) Chronic resveratrol treatment ameliorates cell adhesion and mitigates the inflammatory phenotype in senescent human fibroblasts. J. Gerontol. A Biol. Sci. Med. Sci. 68, 371–381.
  91. Lim, H. et al. (2015) Effects of flavonoids on senescence-associated secretory phenotype formation from bleomycin-induced senescence in BJ fibroblasts. Biochem. Pharmacol. 96, 337–348.
  92. Toso, A. et al. (2015) Enhancing chemotherapy efficacy by reprogramming the senescence-associated secretory phenotype of prostate tumors: a way to reactivate the antitumor immunity. Oncoimmunology 4, e994380.
  93. Xu, M. et al. (2015) Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 4, e12997.
  94. Liou, C.J. et al. (2014) Oral lovastatin attenuates airway inflammation and mucus secretion in ovalbumin-induced murine model of asthma. Allergy Asthma Immunol. Res. 6, 548–557.
  95. Mian, B.M. et al. (2003) Fully human anti-interleukin 8 antibody inhibits tumor growth in orthotopic bladder cancer xenografts via down-regulation of matrix metalloproteases and nuclear factor-kappaB. Clin. Cancer Res. 9, 3167–3175.
  96. Karkera, J. et al. (2011) The anti-interleukin-6 antibody siltuximab down-regulates genes implicated in tumorigenesis in prostate cancer patients from a Phase I study. Prostate 71, 1455–1465.
  97. Pellegrini, G. et al. (2004) Telomerase activity is sufficient to bypass replicative senescence in human limbal and conjunctival but not corneal keratinocytes. Eur. J. Cell Biol. 83, 691–700.
  98. Abad, M. et al. (2013) Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 502, 340–345.
  99. Sagiv, A. et al. (2013) Granule exocytosis mediates immune surveillance of senescent cells. Oncogene 32, 1971–1977.
  100. Demaria, M. et al. (2014) An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733.
  101. Pei, Ming. "Environmental preconditioning rejuvenates adult stem cells' proliferation and chondrogenic potential." Biomaterials (2016).
  102. Vañó-Galván, S., and F. Camacho. "New Treatments for Hair Loss." Actas Dermo-Sifiliográficas (English Edition) (2017).
  103. Anitua, Eduardo, Ander Pino, and Gorka Orive. "Opening new horizons in regenerative dermatology using platelet‐based autologous therapies." International journal of dermatology 56.3 (2017): 247-251.

References and Literature - Stem Cell-based Therapies and Osteoarthritis

  1. Zeckser, J., Wolff, M., Tucker, J., & Goodwin, J. (2016). Multipotent Mesenchymal Stem Cell Treatment for Discogenic Low Back Pain and Disc Degeneration, 2016, 4–6.
  2. Baglio, Serena Rubina, Dirk Michiel Pegtel, and Nicola Baldini. "Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy." Frontiers in physiology 3 (2012): 359.
  3. Richardson, Stephen M., et al. "Mesenchymal stem cells in regenerative medicine: focus on articular cartilage and intervertebral disc regeneration." Methods 99 (2016): 69-80.
  4. Strassburg, Sandra, et al. "Bi-directional exchange of membrane components occurs during co-culture of mesenchymal stem cells and nucleus pulposus cells." PLoS One 7.3 (2012): e33739.
  5. K. A. Pettine, M. B. Murphy, R. K. Suzuki, and T. T. Sand, “Percutaneous lumbar intradiscal injection of autologous bone marrowconcentrated cells significantly reduces discogenic pain through 12 months,” Stem Cells, vol. 33, no. 1, pp. 146–156, 2015.
  6. Mesoblast Reports Positive 24 Month Results in Phase 2 Trial for Chronic Low Back Pain and Initiation of the Phase 3 Program at JP Morgan Healthcare Conference, January 2015, http://www.mesoblast.com. [7] N. Bogduk, C. Aprill, and R. Derby, “Lumbar discogenic pain: state-of-the-art review,” Pain Medicine, vol. 14, no. 6, pp. 813– 836, 2013.
  7. M. J. DePalma, J. M. Ketchum, and T. R. Saullo, “Etiology of chronic low back pain in patients having undergone lumbar fusion,” Pain Medicine, vol. 12, no. 5, pp. 732–739, 2011.
  8. J. C. Iatridis, S. B. Nicoll, A. J. Michalek, B. A. Walter, and M. S. Gupta, “Role of biomechanics in intervertebral disc degeneration and regenerative therapies: what needs repairing in the disc and what are promising biomaterials for its repair?” Spine Journal, vol. 13, no. 3, pp. 243–262, 2013.
  9. N. Bogduk, Practice Guidelines for Spinal Diagnostic and Treatment Procedures, International Spine Intervention Society, San Diego, Calif, USA, 2nd edition, 2013.
  10. K. M.Malik, S. P.Cohen,D. R.Walega, and H. T. Benzon, “Diagnostic criteria and treatment of discogenic pain: a systematic review of recent clinical literature,” The Spine Journal, vol. 13, no. 11, pp. 1675–1689, 2013.
  11. U. G. Longo, N. Papapietro, S. Petrillo, E. Franceschetti, N. Maffulli, and V.Denaro, “Mesenchymal stem cell for prevention and management of intervertebral disc degeneration,” Stem Cells International, vol. 2012, Article ID 921053, 7 pages, 2012.
  12. H. T. J. Gilbert, J. A. Hoyland, and S. M. Richardson, “Stem cell regeneration of degenerated intervertebral discs: current status (Update),” Current Pain and Headache Reports, vol. 17, article 377, 2013.
  13. H. J. Braun, N. Wilcox-Fogel, H. J. Kim, M. A. Pouliot, A. H. S. Harris, and J. L. Dragoo, “The effect of local anesthetic and corticosteroid combinations on chondrocyte viability,” Knee Surgery, Sports Traumatology, Arthroscopy, vol. 20, no. 9, pp. 1689–1695, 2012.
  14. J. L. Dragoo, C. M. Danial, H. J. Braun, M. A. Pouliot, and H. J. Kim, “The chondrotoxicity of single-dose corticosteroids,” Knee Surgery, Sports Traumatology, Arthroscopy, vol. 20, no. 9, pp. 1809–1814, 2012.
  15. B. Peng, X. Pang, Y.Wu, C. Zhao, and X. Song, “A randomized placebo-controlled trial of intradiscal methylene blue injection for the treatment of chronic discogenic lowback pain,” Pain, vol. 149, no. 1, pp. 124–129, 2010.
  16. S.-H. Kim, S.-H. Ahn, Y.-W. Cho, and D.-G. Lee, “Effect of intradiscal methylene blue injection for the chronic discogenic low back pain: one year prospective follow-up study,” Annals of Rehabilitation Medicine, vol. 36, no. 5, pp. 657–664, 2012.
  17. W. Yin, K. Pauza, W. J. Olan, J. F. Doerzbacher, and K. J. Thorne, “Intradiscal injection of fibrin sealant for the treatment of symptomatic lumbar internal disc disruption: results of a prospective multicenter pilot study with 24-month follow-up,” Pain Medicine, vol. 15, no. 1, pp. 16–31, 2014.
  18. S.-Z. Wang, Y.-F. Rui, Q. Tan, and C. Wang, “Enhancing intervertebral disc repair and regeneration through biology: plateletrich plasma as an alternative strategy,” Arthritis Research & Therapy, vol. 15, no. 5, article 220, 2013.
  19. G. B. Gullung, W. Woodall, M. Tucci, J. James, D. Black, and R. McGuire, “Platelet-rich plasma effects on degenerative disc disease: analysis of histology and imaging in an animal model,” Evidence-Based Spine-Care Journal, vol. 2, no. 4, pp. 13–18, 2011.
  20. Y.A.Tuakli-Wosornu, A. Terry, K. Boachie-Adjei et al., “Lumbar intradiskal platelet-rich plasma (PRP) injections: a prospective, double-blind, randomized controlled study,” PM&R, 2015.
  21. L. Manchikanti, F. J. E. Falco, R. M. Benyamin et al., “An update of the systematic assessment of mechanical lumbar disc decompression with nucleoplasty,” Pain Physician, vol. 16, no. 2, pp. SE25–SE54, 2013.
  22. V. Singh, L. Manchikanti, A. K. Calodney et al., “Percutaneous lumbar laser disc decompression: an update of current evidence,” Pain Physician, vol. 16, no. 2, pp. 229–260, 2013.
  23. D. H. Jo and H. J. Yang, “The survey of the patient received the epiduroscopic laser neural decompression,” The Korean Journal of Pain, vol. 26, no. 1, pp. 27–31, 2013.
  24. W. C. H. Jacobs, S. M. Rubinstein, P. C. Willems et al., “The evidence on surgical interventions for low back disorders, an overviewof systematic reviews,” European Spine Journal, vol. 22, no. 9, pp. 1936–1949, 2013.
  25. W. Shohei, Y. Kuroda, F. Ogura, T. Shigemoto, and M. Dezawa, “Regenerative effects of mesenchymal stem cells: contribution of muse cells, a novel pluripotent stem cell type that resides in mesenchymal cells,” Cells, vol. 1, no. 4, pp. 1045–1060, 2012.
  26. P. Ghosh, R.Moore, B. Vernon-Roberts et al., “Immunoselected STRO-3+ mesenchymal precursor cells and restoration of the extracellular matrix of degenerate intervertebral discs: laboratory investigation,” Journal of Neurosurgery: Spine, vol. 16, no. 5, pp. 479–488, 2012.
  27. C. J. Centeno, “Clinical challenges and opportunities of mesenchymal stem cells in musculoskeletal medicine,” PM&R, vol. 6, no. 1, pp. 70–77, 2014.
  28. A. Cond´e-Green, R. L. Rodriguez, S. Slezak, D. P. Singh, N. H. Goldberg, and J. Mclenithan, “Comparison between stromal vascular cells’ isolation with enzymatic digestion and mechanical processing of aspirated adipose tissue,” Plastic and Reconstructive Surgery, vol. 134, no. 4, p. 54, 2014.
  29. L.-L. Lu, Y.-J. Liu, S.-G. Yang et al., “Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis-supportive function and other potentials,” Haematologica, vol. 91, no. 8, pp. 1017–1026, 2006.
  30. H. Brisby, N. Papadimitriou, C. Brantsing, P. Bergh, A. Lindahl, and H. Barreto Henriksson, “The presence of local mesenchymal progenitor cells in human degenerated intervertebral discs and possibilities to influence these in vitro: a descriptive study in humans,” Stem Cells and Development, vol. 22, no. 5, pp. 804– 814, 2013.
  31. M. Tanaka, D. Sakai, A. Hiyama et al., “Effect of cryopreservation on canine and human activated nucleus pulposus cells: a feasibility study for cell therapy of the intervertebral disc,” BioResearch Open Access, vol. 2, no. 4, pp. 273–282, 2013.
  32. D. Coric, K. Pettine, A. Sumich, and M. O. Boltes, “Prospective study of disc repair with allogeneic chondrocytes,” Journal of Neurosurgery: Spine, vol. 18, no. 1, pp. 85–95, 2013.
  33. H. J.Meisel, V. Siodla, T. Ganey, Y.Minkus, W. C. Hutton, and O. J. Alasevic, “Clinical experience in cell-based therapeutics: disc chondrocyte transplantation: a treatment for degenerated or damaged intervertebral disc,” Biomolecular Engineering, vol. 24, no. 1, pp. 5–21, 2007.
  34. T. Miyamoto, T. Muneta, T. Tabuchi et al., “Intradiscal transplantation of synovial mesenchymal stem cells prevents intervertebral disc degeneration through suppression of matrix metalloproteinase-related genes in nucleus pulposus cells in rabbits,”Arthritis Research&Therapy, vol. 12,no. 6, articleR206, 2010.
  35. S. Strassburg, S. M. Richardson, A. J. Freemont, and J. A. Hoyland, “Co-culture induces mesenchymal stem cell differentiation and modulation of the degenerate human nucleus pulposus cell phenotype,” Regenerative Medicine, vol. 5, no. 5, pp. 701–711, 2010.
  36. Z. Sun, Z.-H. Liu, X.-H. Zhao et al., “Impact of direct cell cocultures on human adipose-derived stromal cells and nucleus pulposus cells,” Journal of Orthopaedic Research, vol. 31, no. 11, pp. 1804–1813, 2013.
  37. X. Li, J. P. Lee, G. Balian, and D. G. Anderson, “Modulation of chondrocytic properties of fat-derivedmesenchymal cells in cocultures with nucleus pulposus,”Connective Tissue Research, vol. 46, no. 2, pp. 75–82, 2005.
  38. C. LeVisage, S.W.Kim,K.Tateno, A.N. Sieber, J.P.Kostuik, and K. W. Leong, “Interaction of human mesenchymal stem cells with disc cells: changes in extracellular matrix biosynthesis,” Spine, vol. 31, no. 18, pp. 2036–2042, 2006.
  39. A.W. H. Gebraad, S. Miettinen, D.W. Grijpma, and S. P.Haimi, “Human adipose stem cells in chondrogenic differentiation medium without growth factors differentiate towards annulus fibrosus phenotype in vitro,” Macromolecular Symposia, vol. 334, no. 1, pp. 49–56, 2013.
  40. K.Wuertz, K. Godburn, C. Neidlinger-Wilke, J. Urban, and J. C. Iatridis, “Behavior of mesenchymal stem cells in the chemical microenvironment of the intervertebral disc,” Spine, vol. 33, no. 17, pp. 1843–1849, 2008.
  41. D. Sakai, J. Mochida, T. Iwashina et al., “Regenerative effects of transplanting mesenchymal stem cells embedded in atelocollagen to the degenerated intervertebral disc,” Biomaterials, vol. 27, no. 3, pp. 335–345, 2006.
  42. V. Y. L. Leung, D.M. K. Aladin, F. Lv et al., “Mesenchymal stem cells reduce intervertebral disc fibrosis and facilitate repair,” Stem Cells, vol. 32, no. 8, pp. 2164–2177, 2014.
  43. L. Orozco, R. Soler, C. Morera, M. Alberca, A. S´anchez, and J. Garc´ıa-Sancho, “Intervertebral disc repair by autologous mesenchymal bonemarrowcells: a pilot study,” Transplantation, vol. 92, no. 7, pp. 822–828, 2011.
  44. Mesoblast Limited, “Positive Spinal Disc Repair Trial Results Using Mesoblast Adult Stem Cells,” 2014, http://www.globenewswire. com/.
  45. A. Colombini, C. Ceriani, G. Banfi, M. Brayda-Bruno, and M. Moretti, “Fibrin in intervertebral disc tissue engineering,” Tissue Engineering Part B: Reviews, vol. 20, no. 6, pp. 713–721, 2014.
  46. B. R.Whatley and X.Wen, “Intervertebral disc (IVD): structure, degeneration, repair and regeneration,” Materials Science & Engineering C, vol. 32, no. 2, pp. 61–77, 2012.
  47. Y. Wang, Z.-B. Han, Y.-P. Song, and Z. C. Han, “Safety of mesenchymal stem cells for clinical application,” Stem Cells International, vol. 2012,Article ID 652034, 4 pages, 2012.
  48. G.Vadal'a, G. Sowa,M.Hubert, L. G.Gilbertson,V.Denaro, and J. D. Kang, “Mesenchymal stem cells injection in degenerated intervertebral disc: cell leakage may induce osteophyte formation,” Journal of Tissue Engineering and Regenerative Medicine, vol. 6, no. 5, pp. 348–355, 2012.
  49. W.-H. Chen, H.-Y. Liu, W.-C. Lo et al., “Intervertebral disc regeneration in an ex vivo culture system using mesenchymal stem cells and platelet-rich plasma,” Biomaterials, vol. 30, no. 29, pp. 5523–5533, 2009.
  50. Y. Zhu, M. Yuan, H. Y. Meng et al., “Basic science and clinical application of platelet-rich plasma forcartilage defects and osteoarthritis: a review,” Osteoarthritis and Cartilage, vol. 21, no. 11, pp. 1627–1637, 2013.
  51. C. S. Lee,O. A. Burnsed,V. Raghuram, J.Kalisvaart, B.D. Boyan, and Z. Schwartz, “Adipose stem cells can secrete angiogenic factors that inhibit hyaline cartilage regeneration,” Stem Cell Research &Therapy, vol. 3, no. 4, article 35, 2012.
  52. T. E. Foster, B. L. Puskas, B. R. Mandelbaum, M. B. Gerhardt, and S. A. Rodeo, “Platelet-rich plasma: from basic science to clinical applications,” The American Journal of Sports Medicine, vol. 37, no. 11, pp. 2259–2272, 2009.
  53. A. S.Wasterlain, H. J. Braun, A. H. S. Harris, H.-J. Kim, and J. L. Dragoo, “The systemic effects of platelet-rich plasma injection,” The American Journal of Sports Medicine, vol. 41, no. 1, pp. 186– 193, 2013.

References and Literature - Stem Cell-based Therapies and Parkinson's Disease

  1. Riecke J. et al. "A Meta-Analysis of Mesenchymal Stem Cells in Animal Models of Parkinson's Disease." Stem cells and development 24.18 (2015): 2082-2090.
  2. Ahmed HH. et al. "Updates in the pathophysiological mechanisms of Parkinson’s disease: Emerging role of bone marrow mesenchymal stem cells." World journal of stem cells 8.3 (2016): 106.
  3. Equbal Z. and Mukhopadhyay A. "Counting on Mesenchymal Stem Cells: A Hope for Treating Parkinson’s Disease. J Stem Cells Res." Rev & Rep 3.1 (2016): 1022.
  4. Wei X. et al. "Mesenchymal stem cells: a new trend for cell therapy." Acta Pharmacologica Sinica 34.6 (2013): 747-754.
  5. Wang S. et al. "Clinical applications of mesenchymal stem cells." Journal of hematology & oncology 5.1 (2012): 19.
  6. Gu W. et al. "Transplantation of bone marrow mesenchymal stem cells reduces lesion volume and induces axonal regrowth of injured spinal cord." Neuropathology 30.3 (2010): 205-217.
  7. Wilkins A. et al. "Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro." Stem cell research 3.1 (2009): 63-70.

References and Literature - Stem Cell-based Therapies and Osteoarthritis

  1. A. Hejcl, J. Sedy, M. Kapcalova, D.A. Toro, T. Amemori, P. Lesny, K. Likavcanova-Masinova, E. Krumbholcova, M. Pradny, J. Michalek, M. Burian, M. Hajek, P. Jendelova, E. Sykova, HPMA-RGD hydrogels seeded with mesenchymal stem cells improve functional outcome in chronic spinal cord injury, Stem Cells Dev. 19 (2010) 1535e1546.
  2. Anthony, Diana F., and Paul G. Shiels. "Exploiting paracrine mechanisms of tissue regeneration to repair damaged organs." Transplantation research 2.1 (2013): 10.
  3. Wright KT, Masri WE, Osman A, Chowdhury J, Johnson WEB: Concise review: bone marrow for the tre atment of spinal cord injury: mechanisms and clinical implications. Stem Cells 2011, 29: 169 – 178
  4. Quertainmont R, Cantinieaux D, Bot man O, Eid S, Schoenen J, Franzen R: Mesenchymalstemcellgraftimproves recovery after spinal cord injury in adult rats through neurotrophic and pro-angiogenic actions. PLoS One 2012, 7: e39500.
  5. Lamichhane, Tek N., et al. "Emerging roles for extracellular vesicles in tissue engineering and regenerative medicine." Tissue Engineering Part B: Reviews 21.1 (2014): 45-54.
  6. Thuret, Sandrine, Lawrence DF Moon, and Fred H. Gage. "Therapeutic interventions after spinal cord injury." Nature Reviews Neuroscience 7.8 (2006): 628-643.
  7. Thuret, Sandrine, Lawrence DF Moon, and Fred H. Gage. "Therapeutic interventions after spinal cord injury." Nature Reviews Neuroscience 7.8 (2006): 628-643.
  8. Lindvall O, Kokaia Z: Stem cells for the treatment of neurological disorders. Nature 2006, 441(7097):1094-1096.
  9. Yoon SH, Shim YS, Park YH, Chung JK, Nam JH, Kim MO, Park HC, Park SR, Min BH, Kim EY, et al: Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage-colony stimulating factor: Phase I/II clinical trial. Stem Cells 2007, 25(8):2066-2073.
  10. Karamouzian S, Nematollahi-Mahani SN, Nakhaee N et al (2012) Clinical safety and primary efficacy of bone marrow mesenchymal cell transplantation in subacute spinal cord injured patients. Clin Neurol Neurosurg 114(7):935–939
  11. Saito F, Nakatani T, Iwase M et al (2008) Spinal cord injury treatment with intrathecal autologous bone marrow stromal cell transplantation: the first clinical trial case report. J Trauma 64(1):53–59
  12. Saito F, Nakatani T, Iwase M et al (2012) Administration of cul¬tured autologous bone marrow stromal cells into cerebrospinal fluid in spinal injury patients: a pilot study. Restor Neurol Neu¬rosci 30(2):127–136

References and Literature - Stem Cell-based Therapies and Erectily dysfunction and impotence

  1.  Feldman HA, Goldstein I, Hatzichristou DG, Krane RJ, McKinlay JB. Impotence and its medical and psychosocial correlates: results of the Massachusetts Male Aging Study. J Urol. 1994;151:54–61.
  2. Litwin MS, Nied RJ, Dhanani N. Health-related quality of life in men with erectile dysfunction. J Gen Intern Med. 1998;13:159–166.
  3. Chowdhury SH, Cozma AI, Chowdhury JH. Erectile Dysfunction. Essentials for the Canadian Medical Liscensing Exam: Review and Prep for MCCQE Part I. 2nd edition. Wolters Kluwer. Hong Kong. 2017.
  4. Lue TF, Lee KL. Pharmacotherapy for erectile dysfunction. Chin Med J (Engl) 2000;113:291–298.
  5. Dorsey P, Keel C, Klavens M, Hellstrom WJ. Phosphodiesterase type 5 (PDE5) inhibitors for the treatment of erectile dysfunction. Expert Opin Pharmacother. 2010;11:1109–1122.
    Alwaal A, Hussein AA, Lin CS, Lue TF. Prospects of stem cell treatment in benign urological diseases. Korean J Urol. 2015;56:257–265.
  6. Soebadi MA, Moris L, Castiglione F, Weyne E, Albersen M. Advances in stem cell research for the treatment of male sexual dysfunctions. Curr Opin Urol. 2016;26:129–139.
  7. Gur, Serap, et al. "Advances in stem cell therapy for erectile dysfunction." Expert opinion on biological therapy just-accepted (2018).
  8. Panda, Arabind. "Stem cell in urology—are we at the cusp of a new era?." Translational Andrology and Urology (2018).
  9. https://en.wikipedia.org/wiki/Erection Anatomy & Physiology, Connexions Web site. http://cnx.org/content/col11496/1.6/, Jun 19, 2013.
  10. Lue TF. Erectile dysfunction. N Engl J Med. 2000;342:1802–1813.
  11. Prieto D. Physiological regulation of penile arteries and veins. Int J Impot Res. 2008;20:17–29.
  12. Lin CS, Xin ZC, Wang Z, et al. Stem cell therapy for erectile dysfunction: a critical review. Stem Cells Dev. 2012;21:343–351.
  13. Mulhall JP, Bella AJ, Briganti A, et al. Erectile function rehabilitation in the radical prostatectomy patient. J Sex Med. 2010;7:1687–1698.
  14. Iacono F, Giannella R, Somma P, Manno G, Fusco F, Mirone V. Histological alterations in cavernous tissue after radical prostatectomy. J Urol. 2005;173:1673–1676.
  15. Fode M, Ohl DA, et al. Penile rehabilitation after radical prostatectomy: what the evidence really says. BJU Int. 2013;112:998–1008.
  16. Dashwood MR, Crump A, Shi-Wen X, et al. Identification of neuronal nitric oxide synthase (nNOS) in human penis: a potential role of reduced neuronally-derived nitric oxide in erectile dysfunction. Curr Pharm Biotechnol. 2011;12:1316–1321.
  17. Gratzke C, Angulo J, Chitaley K, et al. Anatomy, physiology, and pathophysiology of erectile dysfunction. J Sex Med. 2010;7:445–475.
  18. Huang YC, Ning H, Shindel AW, et al. The effect of intracavernous injection of adipose tissue-derived stem cells on hyperlipidemia-associated erectile dysfunction in a rat model. J Sex Med. 2010;7:1391–1400.
  19. Chen S, Liu Z, Tian N, et al. Intracoronary transplantation of autologous bone marrow mesenchymal stem cells for ischemic cardiomyopathy due to isolated chronic occluded left anterior descending artery. J Invasive Cardiol. 2006;18:552–556.
  20. Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57:874–882.
  21. Pal R, Venkataramana NK, Bansal A, et al. Ex vivo-expanded autologous bone marrow-derived mesenchymal stromal cells in human spinal cord injury/paraplegia: a pilot clinical study. Cytotherapy. 2009;11:897–911.
  22. Venkataramana NK, Kumar SK, Balaraju S, et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson's disease. Transl Res. 2010;155:62–70.
  23. Violaine K. Harris, James Stark, Tamara Vyshkina, et al. Phase I Trial of Intrathecal Mesenchymal Stem Cell-derived Neural Progenitors in Progressive Multiple Sclerosis. EBioMedicine by The Lancet. March 2018 Volume 29, Pages 23–30.
  24. Sun L, Akiyama K, Zhang H, et al. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. Stem Cells. 2009;27:1421–1432.
  25. Dash NR, Dash SN, Routray P, et al. Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow-derived mesenchymal stem cells. Rejuvenation Res. 2009;12:359–366.
  26. Wakitani S, Okabe T, Horibe S, et al. Safety of autologous bone marrow-derived mesenchymal stem cell transplantation for cartilage repair in 41 patients with 45 joints followed for up to 11 years and 5 months. J Tissue Eng Regen Med. 2011;5:146–150.
  27. Khera M, Albersen M, et al. Mesenchymal stem cell therapy for the treatment of erectile dysfunction. J Sex Med. 2015;12:1105–1106.
  28. Soebadi MA, Moris L, Castiglione F, et al. Advances in stem cell research for the treatment of male sexual dysfunctions. Curr Opin Urol. 2016;26:129–139.
  29. Zhang H, Yang R, Wang Z, et al. Adipose tissue-derived stem cells secrete CXCL5 cytokine with neurotrophic effects on cavernous nerve regeneration. J Sex Med. 2011;8:437–446.
  30. Albersen M, Lin CS, Lue T. Stem-cell therapy for erectile dysfunction. Arab J Urol. 2013;11:237–244.
  31. Qiu X, Villalta J, Ferretti L, et al. Effects of intravenous injection of adipose-derived stem cells in a rat model of radiation therapy-induced erectile dysfunction. J Sex Med. 2012;9:1834–1841.
  32. Alwaal A, Hussein AA, Lin CS, Lue TF. Prospects of stem cell treatment in benign urological diseases. Korean J Urol. 2015;56:257–265.
  33. You D, Jang MJ, Lee J, et al. Periprostatic implantation of human bone marrow-derived mesenchymal stem cells potentiates recovery of erectile function by intracavernosal injection in a rat model of cavernous nerve injury. Urology. 2013;81:104–110.
  34. You D, Jang MJ, Lee J, et al. Comparative analysis of periprostatic implantation and intracavernosal injection of human adipose tissue-derived stem cells for erectile function recovery in a rat model of cavernous nerve injury. Prostate. 2013;73:278–286.
  35. Lin CS, Xin Z, Dai J, Huang YC, Lue TF. Stem-cell therapy for erectile dysfunction. Expert Opin Biol Ther. 2013;13:1585–1597.
  36. Lin G, Yang R, Banie L, et al. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate. 2010;70:1066–1073.
  37. Bahk JY, Jung JH, Han H, et al. Treatment of diabetic impotence with umbilical cord blood stem cell intracavernosal transplant: preliminary report of 7 cases. Exp Clin Transplant. 2010;8:150–160.
  38. Yiou R, Hamidou L, Birebent B, et al. Safety of intracavernous bone marrow-mononuclear cells for postradical prostatectomy erectile dysfunction: an open dose-escalation pilot study. Eur Urol. 2016;69:988–991.
  39. Haahr MK, Jensen CH, Toyserkani NM, et al. Safety and potential effect of a single intracavernous injection of autologous adipose-derived regenerative cells in patients with erectile dysfunction following radical prostatectomy: an open-label phase I clinical trial. EBioMedicine. 2016;5:204–210.
  40. Levy JA, Marchand M, Iorio L, et al. Determining the feasibility of managing erectile dysfunction in humans with placental-derived stem cells. J Am Osteopath Assoc. 2016;116:e1–e5.
  41. Fandel TM, Albersen M, Lin G, et al. Recruitment of intracavernously injected adipose-derived stem cells to the major pelvic ganglion improves erectile function in a rat model of cavernous nerve injury. Eur Urol. 2012;61:201–210.
  42. Takayanagi A, Sasaki M, Kataoka-Sasaki Y, et al. Intravenous preload of mesenchymal stem cells rescues erectile function in a rat model of cavernous nerve injury. J Sex Med. 2015;12:1713–1721.
  43. Vakalopoulos, Ioannis, et al. "Stem cell therapy in erectile dysfunction: science fiction or realistic treatment option?." Hormones (2018): 1-6.
  44. Wu, Han, et al. "Nanotechnology-assisted adipose-derived stem cell (ADSC) therapy for erectile dysfunction of cavernous nerve injury: In vivo cell tracking, optimized injection dosage, and functional evaluation." Asian journal of andrology 20.5 (2018): 442.
  45. Matz, Ethan L., et al. "Stem Cell Therapy for Erectile Dysfunction." Sexual medicine reviews (2018).
  46. Shan, H., Chen, F., Zhang, T., He, S., Xu, L., & Wei, A. (2015). Stem cell therapy for erectile dysfunction of cavernous nerve injury rats: A systematic review and meta-analysis. PLoS ONE, 10(4), 1–23. http://doi.org/10.1371/journal.pone.0121428
  47. You, Dalsan, et al. "Comparative Study of Autologous Stromal Vascular Fraction and Adipose‐Derived Stem Cells for Erectile Function Recovery in a Rat Model of Cavernous Nerve Injury." Stem cells translational medicine 4.4 (2015): 351-358.
  48. Chen, X., Yang, Q., Zheng, T., Bian, J., Sun, X., Shi, Y., … Deng, C. (2016). Neurotrophic Effect of Adipose Tissue-Derived Stem Cells on Erectile Function Recovery by Pigment Epithelium-Derived Factor Secretion in a Rat Model of Cavernous Nerve Injury, 2016.
  49. Yiou, R., Hamidou, L., Birebent, B., Bitari, D., Lecorvoisier, P., Contremoulins, I., … Rouard, H. (2016). Safety of Intracavernous Bone Marrow-Mononuclear Cells for Postradical Prostatectomy Erectile Dysfunction: An Open Dose-Escalation Pilot Study. European Urology, 69(6), 988–991. http://doi.org/10.1016/j.eururo.2015.09.026
  50. Wang, X. Y., Liu, C. L., Li, S. D., Xu, Y., Chen, P., Liu, Y., … Yang, M. H. (2015). Hypoxia precondition promotes adipose-derived mesenchymal stem cells based repair of diabetic erectile dysfunction via augmenting angiogenesis and neuroprotection. PLoS ONE, 10(3), 1–18. http://doi.org/10.1371/journal.pone.0118951
  51. Haahr, M. K., Jensen, C. H., Toyserkani, N. M., Andersen, D. C., Damkier, P., S??rensen, J. A., … Sheikh, S. P. (2016). Safety and Potential Effect of a Single Intracavernous Injection of Autologous Adipose-Derived Regenerative Cells in Patients with Erectile Dysfunction Following Radical Prostatectomy: An Open-Label Phase I Clinical Trial. EBioMedicine, 5, 204–210. http://doi.org/10.1016/j.ebiom.2016.01.024
  52. Bochinski D, Lin GT, Nunes L, Carrion R, Rahman N, Lin CS, et al. (2004) The effect of neural embryonic stem cell therapy in a rat model of cavernosal nerve injury. Bju Int 94: 904–09. PMID: 15476533
  53. Kim Y, de Miguel F, Usiene I, Rahman N, Yoshimura N, Huard J, et al. (2006) Injection of skeletal muscle-derived cells into the penis improves erectile function. Int J Impot Res 18: 329–34. PMID: 16341028
  54. Fall PA, Izikki M, Tu L, Swieb S, Giuliano F, Bernabe J, et al. (2009) Apoptosis and effects of intracavernous bone marrow cell injection in a rat model of postprostatectomy erectile dysfunction. Eur Urol. 2009; 56: 716–25. doi: 10.1016/j.eururo.2008.09.059 PMID: 18922625
  55. Albersen M, Fandel TM, Lin G, Wang G, Banie L, Lin CS, et al. (2010) Injections of adipose tissue-derived stem cells and stem cell lysate improve recovery of erectile function in a rat model of cavernous nerve injury. J Sex Med 7: 3331–40. doi: 10.1111/j.1743-6109.2010.01875.x PMID: 20561166
  56. Kendirci M, Trost L, Bakondi B, Whitney MJ, Hellstrom WJ, Spees JL (2010) Transplantation of nonhematopoietic adult bone marrow stem/progenitor cells isolated by p75 nerve growth factor receptor into the penis rescues erectile function in a rat model of cavernous nerve injury. J Urol 184: 1560–66. doi: 10.1016/j.juro.2010.05.088 PMID: 20728109
  57. Lin G, Qiu X, Fandel T, Banie L, Wang G, Lue TF, et al. (2011) Tracking intracavernously injected adipose-derived stem cells to bone marrow. Int J Impot Res 23: 268–75. doi: 10.1038/ijir.2011.38 PMID: 21796145
  58. Lin G, Albersen M, Harraz AM, Fandel TM, Garcia M, McGrath MH, et al. (2011) Cavernous nerve repair with allogenic adipose matrix and autologous adipose-derived stem cells. Urology 77: 1501–09. Woo JC, Bae WJ, Kim SJ, Kim SD, Sohn DW, Hong SH, et al. (2011) Transplantation of muscle-derived stem cells into the corpus cavernosum restores erectile function in a rat model of cavernous nerve injury.
  59. Korean J Urol 52: 359–63. doi: 10.4111/kju.2011.52.5.359 PMID: 21687398 Fandel TM, Albersen M, Lin G, Qiu X, Ning H, Banie L, et al. (2012) Recruitment of intracavernously injected adipose-derived stem cells to the major pelvic ganglion improves erectile function in a rat model of cavernous nerve injury. Eur Urol 61: 201–10. doi: 10.1016/j.eururo.2011.07.061 PMID: 21824718
  60. Kim SJ, Park SH, Sung YC, Kim SW (2012) Effect of mesenchymal stem associated to matrixen on the erectile function in the rat model with bilateral cavernous nerve crushing injury. Int Braz J Urol 38: 833– 41. PMID: 23302404
  61. Kim SJ, Choi SW, Hur KJ, Park SH, Sung YC, Ha YS, et al. (2012) Synergistic effect of mesenchymal stem cells infected with recombinant adenovirus expressing human BDNF on erectile function in a rat model of cavernous nerve injury. Korean J Urol 53: 726–32. doi: 10.4111/kju.2012.53.10.726 PMID: 23136635
  62. Kovanecz I, Rivera S, Nolazco G, Vernet D, Segura D, Gharib S, et al. (2012) Separate or combined treatments with daily sildenafil, molsidomine, or muscle-derived stem cells prevent erectile dysfunction in a rat model of cavernosal nerve damage. J Sex Med 9: 2814–26. doi: 10.1111/j.1743-6109.2012. 02913.x PMID: 22974131
  63. Piao S, Kim IG, Lee JY, Hong SH, Kim SW, Hwang TK, et al. (2012) Therapeutic effect of adipose-derived stem cells and BDNF-immobilized PLGA membrane in a rat model of cavernous nerve injury. J Sex Med 9: 1968–79. doi: 10.1111/j.1743-6109.2012.02760.x PMID: 22642440
  64. Qiu X, Villalta J, Ferretti L, Fandel TM, Albersen M, Lin G, et al. (2012) Effects of intravenous injection of adipose-derived stem cells in a rat model of radiation therapy-induced erectile dysfunction. J Sex Med 9: 1834–41. doi: 10.1111/j.1743-6109.2012.02753.x PMID: 22548750
  65. Albersen M, Fandel TM, Lin G, Wang G, Banie L, Lin CS, et al. (2010) Injections of adipose tissue-derived stem cells and stem cell lysate improve recovery of erectile function in a rat model of cavernous nerve injury. J Sex Med 7: 3331–40. doi: 10.1111/j.1743-6109.2010.01875.x PMID: 20561166
  66. Tewari, A., et al. "Technique of traction-free nerve-sparing robotic prostatectomy: delicate tissue handling by real-time penile oxygen monitoring." International journal of impotence research 24.1 (2012): 11.

References and Literature - Stem Cell-based Therapies for Rheumatoid Arthritis

  1. Tyndall A, van Laar JM. Stem cells in the treatment of inflammatory arthritis. Best Pract Res Clin Rheumatol 2010;24:565–574.
  2. Liang J, Li X, Zhang H, Wang D, Feng X, Wang H, et al. Allogeneic mesenchymal stem cells transplantation in patients with refractory RA. Clin Rheumatol 2012;31:157–161.
  3. Wang L, Wang L, Cong X, Liu G, Zhou J, Bai B, et al. Human umbilical cord mesenchymal stem cell therapy for patients with active rheumatoid arthritis: safety and efficacy. Stem Cells Dev 2013;22:3192–3202.

  1. Georg Hansmann, Philippe Chouvarine, Franziska Diekmann, Martin Giera, Markus Ralser, Michael Mülleder, Constantin von Kaisenberg, Harald Bertram, Ekaterina Legchenko & Ralf Hass "Human umbilical cord mesenchymal stem cell-derived treatment of severe pulmonary arterial hypertension". Nature Cardiovascular Research volume 1, pages568–576 (2022).
  2. Murphy JM, Fink DJ, Hunziker EB, et al. Stem cell therapy in a caprine model of osteoarthritis . Arthritis Rheum. 2003;48:3464–74.
  3. Lee KB, Hui JH, Song IC, Ardany L, et al. Injectable mesenchymal stem cell therapy for large cartilage defects—a porcine model. Stem Cell. 2007;25:2964–71.
  4. Saw KY, Hussin P, Loke SC, et al. Articular cartilage regeneration with autologous marrow aspirate and hyaluronic acid: an experimental study in a goat model. Arthroscopy . 2009;25(12):1391–400.
  5. Black L, Gaynor J, Adams C, et al. Effect of intra-articular injection of autologous adipose-derived mesenchymal stem and regenerative cells on clinical signs of chronic osteoarthritis of the elbow joint in dogs. Vet Ther. 2008;9:192-200.
  6. Centeno C, Busse D, Kisiday J, et al. Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells. Pain Physician. 2008;11(3):343–53.
  7. Centeno C, Kisiday J, Freeman M, et al. Partial regeneration of the human hip via autologous bone marrow nucleated cell transfer: a case study. Pain Physician. 2006;9:253–6.
  8. Centeno C, Schultz J, Cheever M. Safety and complications reporting on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell. 2011;5(1):81–93.
  9. Pak J. Regeneration of human bones in hip osteonecrosis and human cartilage in knee osteoarthritis with autologous adipose derived stem cells: a case series. J Med Case Rep. 2001;5:296.
  10. Kuroda R, Ishida K, et al. Treatment of a full-thickness articular cartilage defect in the femoral condyle of an athlete with autologous bone-marrow stromal cells. Osteoarthritis Cartilage. 2007;15:226–31.
  11. Emadedin M, Aghdami N, Taghiyar L, et al. Intra-articular injection of autologous mesenchymal stem cells in six patients with knee osteoarthritis. Arch Iran Med. 2012;15(7):422–8.
  12. Saw KY et al. Articular cartilage regeneration with autologous peripheral blood stem cells versus hyaluronic acid: a randomized controlled trial. Arthroscopy. 2013;29(4):684–94.
  13. Vangsness CT, Farr J, Boyd J, et al. Adult human mesenchymal stem cells delivered via intra-articular injection to the knee following partial medial meniscectomy. J Bone Joint Surg. 2014;96(2):90–8.
  14. Freitag, Julien, et al. Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy–a review. BMC musculoskeletal disorders 17.1 (2016): 230.
  15. Maumus, Marie, Christian Jorgensen, and Danièle Noël. " Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes. " Biochimie 95.12 (2013): 2229-2234.
  16. Dostert, Gabriel, et al. " How do mesenchymal stem cells influence or are influenced by microenvironment through extracellular vesicles communication?. " Frontiers in Cell and Developmental Biology 5 (2017).
  17. Chaparro, Orlando, and Itali Linero. " Regenerative Medicine: A New Paradigm in Bone Regeneration. " (2016).
  18. Toh, Wei Seong, et al. " MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment. " Seminars in Cell & Developmental Biology. Academic Press, 2016.
  19. Chaparro, Orlando, and Itali Linero. " Regenerative Medicine: A New Paradigm in Bone Regeneration. " (2016).
  20. S. Koelling, J. Kruegel, M. Irmer, J.R. Path, B. Sadowski, X. Miro, et al., Migratory chondrogenic progenitor cells from repair tissue during the later stages of human osteoarthritis , Cell Stem Cell 4 (2009) 324–335.
  21. B.A. Jones, M. Pei, Synovium-Derived stem cells: a tissue-Specific stem cell for cartilage engineering and regeneration , Tissue Eng. B: Rev. 18 (2012) 301–311.
  22. W. Ando, J.J. Kutcher, R. Krawetz, A. Sen, N. Nakamura, C.B. Frank, et al., Clonal analysis of synovial fluid stem cells to characterize and identify stable mesenchymal stromal cell/mesenchymal progenitor cell phenotypes in a porcine model: a cell source with enhanced commitment to the chondrogenic lineage, Cytotherapy 16 (2014) 776–788.
  23. K.B.L. Lee, J.H.P. Hui, I.C. Song, L. Ardany, E.H. Lee, Injectable mesenchymal stem cell therapy for large cartilage defects—a porcine model, Stem Cells 25 (2007) 2964–2971.
  24. W.-L. Fu, C.-Y. Zhou, J.-K. Yu, A new source of mesenchymal stem cells for articular cartilage repair: mSCs derived from mobilized peripheral blood share similar biological characteristics in vitro and chondrogenesis in vivo as MSCs from bone marrow in a rabbit model , Am. J. Sports Med. 42 (2014) 592–601.
  25. X. Xie, Y. Wang, C. Zhao, S. Guo, S. Liu, W. Jia, et al., Comparative evaluation of MSCs from bone marrow and adipose tissue seeded in PRP-derived scaffold for cartilage regeneration , Biomaterials 33 (2012) 7008–7018.
  26. E.-R. Chiang, H.-L. Ma, J.-P. Wang, C.-L. Liu, T.-H. Chen, S.-C. Hung, Allogeneic mesenchymal stem cells in combination with hyaluronic acid for the treatment of osteoarthritis in rabbits , PLoS One 11 (2016) e0149835.
  27. H. Nejadnik, J.H. Hui, E.P. Feng Choong, B.-C. Tai, E.H. Lee, Autologous bone marrow–derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study , Am. J. Sports Med. 38 (2010) 1110–1116.
  28. I. Sekiya, T. Muneta, M. Horie, H. Koga, Arthroscopic transplantation of synovial stem cells improves clinical outcomes in knees with cartilage defects , Clin. Orthop. Rel. Res. 473 (2015) 2316–2326.
  29. Y.S. Kim, Y.J. Choi, Y.G. Koh, Mesenchymal stem cell implantation in knee osteoarthritis: an assessment of the factors influencing clinical outcomes , Am. J. Sports Med. 43 (2015) 2293–2301.
  30. W.-L. Fu, Y.-F. Ao, X.-Y. Ke, Z.-Z. Zheng, X. Gong, D. Jiang, et al., Repair of large full-thickness cartilage defect by activating endogenous peripheral blood stem cells and autologous periosteum flap transplantation combined with patellofemoral realignment , Knee 21 (2014) 609–612.
  31. Y.-G. Koh, O.-R. Kwon, Y.-S. Kim, Y.-J. Choi, D.-H. Tak, Adipose-derived mesenchymal stem cells with microfracture versus microfracture alone: 2-year follow-up of a prospective randomized trial , Arthrosc. J. Arthrosc. Relat. Surg. 32 (2016) 97–109.
  32. T.S. de Windt, L.A. Vonk, I.C.M. Slaper-Cortenbach, M.P.H. van den Broek, R. Nizak, M.H.P. van Rijen, et al., Allogeneic mesenchymal stem cells stimulate cartilage regeneration and are safe for single-Stage cartilage repair in humans upon mixture with recycled autologous chondrons , Stem Cells (2016) (n/a-n/a).
  33. L. da Silva Meirelles, A.M. Fontes, D.T. Covas, A.I. Caplan, Mechanisms involved in the therapeutic properties of mesenchymal stem cells , Cytokine Growth Factor Rev. 20 (2009) 419–427.
  34. W.S. Toh, C.B. Foldager, M. Pei, J.H.P. Hui, Advances in mesenchymal stem cell-based strategies for cartilage repair and regeneration , Stem Cell Rev. Rep. 10 (2014) 686–696.
  35. R.C. Lai, F. Arslan, M.M. Lee, N.S.K. Sze, A. Choo, T.S. Chen, et al., Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury , Stem Cell Res. 4 (2010) 214–222.
  36. S. Zhang, W.C. Chu, R.C. Lai, S.K. Lim, J.H.P. Hui, W.S. Toh, Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration, Osteoarthr . Cartil. 24 (2016) 2135–2140.
  37. S. Zhang, W. Chu, R. Lai, J. Hui, E. Lee, S. Lim, et al., 21 – human mesenchymal stem cell-derived exosomes promote orderly cartilage regeneration in an immunocompetent rat osteochondral defect model , Cytotherapy 18 (2016) S13.
  38. C.T. Lim, X. Ren, M.H. Afizah, S. Tarigan-Panjaitan, Z. Yang, Y. Wu, et al., Repair of osteochondral defects with rehydrated freeze-dried oligo[poly(ethylene glycol) fumarate] hydrogels seeded with bone marrow mesenchymal stem cells in a porcine model
  39. A. Gobbi, G. Karnatzikos, S.R. Sankineani, One-step surgery with multipotent stem cells for the treatment of large full-thickness chondral defects of the knee , Am. J. Sports Med. 42 (2014) 648–657.
  40. A. Gobbi, C. Scotti, G. Karnatzikos, A. Mudhigere, M. Castro, G.M. Peretti, One-step surgery with multipotent stem cells and Hyaluronan-based scaffold for the treatment of full-thickness chondral defects of the knee in patients older than 45 years , Knee Surg. Sports Traumatol. Arthrosc. (2016) 1–8.
  41. A. Gobbi, G. Karnatzikos, C. Scotti, V. Mahajan, L. Mazzucco, B. Grigolo, One-step cartilage repair with bone marrow aspirate concentrated cells and collagen matrix in full-thickness knee cartilage lesions: results at 2-Year follow-up , Cartilage 2 (2011) 286–299.
  42. K.L. Wong, K.B.L. Lee, B.C. Tai, P. Law, E.H. Lee, J.H.P. Hui, Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up , Arthrosc. J. Arthrosc. Relat. Surg. 29 (2013) 2020–2028.
  43. J.M. Hare, J.E. Fishman, G. Gerstenblith, et al., Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the poseidon randomized trial, JAMA 308 (2012) 2369–2379.
  44. L. Wu, J.C.H. Leijten, N. Georgi, J.N. Post, C.A. van Blitterswijk, M. Karperien, Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation , Tissue Eng. A 17 (2011) 1425–1436.
  45. L. Wu, H.-J. Prins, M.N. Helder, C.A. van Blitterswijk, M. Karperien, Trophic effects of mesenchymal stem cells in chondrocyte Co-Cultures are independent of culture conditions and cell sources , Tissue Eng. A 18 (2012) 1542–1551.
  46. S.K. Sze, D.P.V. de Kleijn, R.C. Lai, E. Khia Way Tan, H. Zhao, K.S. Yeo, et al., Elucidating the secretion proteome of human embryonic stem cell-derived mesenchymal stem cells , Mol. Cell. Proteomics 6 (2007) 1680–1689.
  47. M.B. Murphy, K. Moncivais, A.I. Caplan, Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine , Exp. Mol. Med. 45 (2013) e54.
  48. M.J. Lee, J. Kim, M.Y. Kim, Y.-S. Bae, S.H. Ryu, T.G. Lee, et al., Proteomic analysis of tumor necrosis factor--induced secretome of human adipose tissue-derived mesenchymal stem cells , J. Proteome Res. 9 (2010) 1754–1762.
  49. S. Bruno, C. Grange, M.C. Deregibus, R.A. Calogero, S. Saviozzi, F. Collino, et al., Mesenchymal stem cell-derived microvesicles protect against acute tubular injury, J. Am. Soc. Nephrol. 20 (2009) 1053–1067.
  50. M. Yá˜nez-Mó, P.R.-M. Siljander, Z. Andreu, A.B. Zavec, F.E. Borràs, E.I. Buzas, et al. Biological properties of extracellular vesicles and their physiological functions (2015).
  51. C. Lawson, J.M. Vicencio, D.M. Yellon, S.M. Davidson, Microvesicles and exosomes: new players in metabolic and cardiovascular disease , J. Endocrinol. 228 (2016) R57–R71.
  52. A.G. Thompson, E. Gray, S.M. Heman-Ackah, I. Mager, K. Talbot, S.E. Andaloussi, et al., Extracellular vesicles in neurodegenerative diseas—pathogenesis to biomarkers, Nat. Rev. Neurol. 12 (2016) 346–357.
  53. I.E.M. Bank, L. Timmers, C.M. Gijsberts, Y.-N. Zhang, A. Mosterd, J.-W. Wang, et al., The diagnostic and prognostic potential of plasma extracellular vesicles for cardiovascular disease , Expert Rev. Mol. Diagn. 15 (2015) 1577–1588.
  54. T. Kato, S. Miyaki, H. Ishitobi, Y. Nakamura, T. Nakasa, M.K. Lotz, et al., Exosomes from IL-1 stimulated synovial fibroblasts induce osteoarthritic changes in articular chondrocytes , Arthritis. Res. Ther. 16 (2014) 1–11.
  55. R.W.Y. Yeo, S.K. Lim, Exosomes and their therapeutic applications, in: C. Gunther, A. Hauser, R. Huss (Eds.), Advances in Pharmaceutical Cell TherapyPrinciples of Cell-Based Biopharmaceuticals, World Scientific, Singapore, 2015, pp. 477–491.
  56. X. Qi, J. Zhang, H. Yuan, Z. Xu, Q. Li, X. Niu, et al., Exosomes secreted by human-Induced pluripotent stem cell-derived mesenchymal stem cells repair critical-sized bone defects through enhanced angiogenesis and osteogenesis in osteoporotic rats , Int. J. Biol. Sci. 12 (2016) 836–849.
  57. R.C. Lai, F. Arslan, S.S. Tan, B. Tan, A. Choo, M.M. Lee, et al., Derivation and characterization of human fetal MSCs: an alternative cell source for large-scale production of cardioprotective microparticles , J. Mol. Cell. Cardiol. 48 (2010) 1215–1224.
  58. Y. Zhou, H. Xu, W. Xu, B. Wang, H. Wu, Y. Tao, et al., Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro , Stem Cell Res. Ther. 4 (2013) 1–13.
  59. Y. Qin, L. Wang, Z. Gao, G. Chen, C. Zhang, Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo , Sci. Rep. 6 (2016) 21961.
  60. M. Nakano, K. Nagaishi, N. Konari, Y. Saito, T. Chikenji, Y. Mizue, et al., Bone marrow-derived mesenchymal stem cells improve diabetes-induced cognitive impairment by exosome transfer into damaged neurons and astrocytes , Sci. Rep. 6 (2016) 24805.
  61. K. Nagaishi, Y. Mizue, T. Chikenji, M. Otani, M. Nakano, N. Konari, et al., Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes , Sci. Rep. 6 (2016) 34842.
  62. S.R. Baglio, K. Rooijers, D. Koppers-Lalic, F.J. Verweij, M. Pérez Lanzón, N. Zini, et al., Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species , Stem Cell Res. Ther. 6 (2015) 1–20.
  63. T. Chen, R. Yeo, F. Arslan, Y. Yin, S. Tan, Efficiency of exosome production correlates inversely with the developmental maturity of MSC donor, J. Stem Cell Res. Ther. 3 (2013) 2.
  64. R.C. Lai, S.S. Tan, B.J. Teh, S.K. Sze, F. Arslan, D.P. de Kleijn, et al., Proteolytic potential of the MSC exosome proteome: implications for an exosome-mediated delivery of therapeutic proteasome , Int. J. Proteomics 2012 (2012) 971907.
  65. T.S. Chen, R.C. Lai, M.M. Lee, A.B.H. Choo, C.N. Lee, S.K. Lim, Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs , Nucleic Acids Res. 38 (2010) 215–224.
  66. R.W. Yeo, R.C. Lai, K.H. Tan, S.K. Lim, Exosome: a novel and safer therapeutic refinement of mesenchymal stem cell, J. Circ. Biomark. 1 (2013) 7.
  67. R.C. Lai, R.W. Yeo, S.K. Lim, Mesenchymal stem cell exosomes, Semin. Cell Dev. Biol. 40 (2015) 82–88.
  68. B. Zhang, R.W. Yeo, K.H. Tan, S.K. Lim, Focus on extracellular vesicles: therapeutic potential of stem cell-derived extracellular vesicles , Int. J. Mol. Sci. 17 (2016) 174.
  69. Hu G-w, Q. Li, X. Niu, B. Hu, J. Liu, Zhou S-m, et al., Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice , Stem Cell Res. Ther. 6 (2015) 1–15.
  70. J. Zhang, J. Guan, X. Niu, G. Hu, S. Guo, Q. Li, et al., Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis , J. Transl. Med. 13 (2015) 1–14.
  71. B. Zhang, M. Wang, A. Gong, X. Zhang, X. Wu, Y. Zhu, et al., HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing, Stem Cells 33 (2015) 2158–2168.
  72. B. Zhang, Y. Yin, R.C. Lai, S.S. Tan, A.B.H. Choo, S.K. Lim, Mesenchymal stem cells secrete immunologically active exosomes , Stem Cells Dev. 23 (2013) 1233–1244.
  73. C.Y. Tan, R.C. Lai, W. Wong, Y.Y. Dan, S.-K. Lim, H.K. Ho, Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models , Stem Cell Res. Ther. 5 (2014) 1–14.
  74. C. Lee, S.A. Mitsialis, M. Aslam, S.H. Vitali, E. Vergadi, G. Konstantinou, et al., Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension , Circulation 126 (2012) 2601–2611.
  75. B. Yu, H. Shao, C. Su, Y. Jiang, X. Chen, L. Bai, et al., Exosomes derived from MSCs ameliorate retinal laser injury partially by inhibition of MCP-1 , Sci. Rep. 6 (2016) 34562.
  76. Jo CH, Lee YG, Shin WH, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof of concept clinical trial. Stem Cells. 2014;32(5):1254–66.
  77. Vega, Aurelio, et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation. 2015;99(8):1681–90.
  78. Davatchi F, Sadeghi-Abdollahi B, Mohyeddin M, et al. Mesenchymal stem cell therapy for knee osteoarthritis. Preliminary report of four patients. Int J Rheum Dis. 2011;14(2):211–5
  79. Hernigou P, Flouzat Lachaniette CH, Delambre J, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case- controlled study. Int Orthop. 2014;38(9):1811–1818
  80. Galli D, Vitale M, Vaccarezza M. Bone marrow-derived mesenchymal cell differentiation toward myogenic lineages: facts and perspectives. Biomed Res Int. 2014;2014:6.
  81. Beitzel K, Solovyova O, Cote MP, et al. The future role of mesenchymal Stem cells in The management of shoulder disorders . Arthroscopy. 2013;29(10):1702–1711.
  82. Isaac C, Gharaibeh B, Witt M, Wright VJ, Huard J. Biologic approaches to enhance rotator cuff healing after injury. J Shoulder Elbow Surg. 2012;21(2):181–190.
  83. Malda, Jos, et al. " Extracellular vesicles [mdash] new tool for joint repair and regeneration. " Nature Reviews Rheumatology (2016).

  1. Xu, Ming, et al. " Transplanted senescent cells induce an osteoarthritis-like condition in mice. " The Journals of Gerontology Series A: Biological Sciences and Medical Sciences (2016): glw154.
  2. McCulloch, Kendal, Gary J. Litherland, and Taranjit Singh Rai. " Cellular senescence in osteoarthritis pathology ." Aging Cell (2017).

Patient Services at ANOVA Institute for Regenerative Medicine

  • Located in the center of Germany, quick access by car or train from anywhere in Europe
  • Simple access worldwide, less than 20 minutes from Frankfurt Airport
  • Individualized therapy with state-of-the-art stem cell products
  • Individually planned diagnostic work-up which include world-class MRI and CT scans
  • German high quality standard on safety and quality assurance
  • Personal service with friendly, dedicated Patient Care Managers
  • Scientific collaborations with academic institutions to assure you the latest regenerative medical programs