全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Advances in Stem Cell Therapy for Erectile Dysfunction

DOI: 10.1155/2014/140618

Full-Text   Cite this paper   Add to My Lib

Abstract:

Stem cell (SC) therapy for erectile dysfunction (ED) has been investigated in 35 published studies, with one being a small-scale clinical trial. Out of these 35 studies, 19 are concerned with cavernous nerve (CN) injury-associated ED while 10 with diabetes mellitus- (DM-) associated ED. Adipose-derived SCs (ADSCs) were employed in 18 studies while bone marrow SCs (BMSCs) in 9. Transplantation of SCs was done mostly by intracavernous (IC) injection, as seen in 25 studies. Allogeneic and xenogeneic transplantations have increasingly been performed but their immune-incompatibility issues were rarely discussed. More recent studies also tend to use combinatory therapies by modifying or supplementing SCs with angiogenic or neurotrophic genes or proteins. All studies reported better erectile function with SC transplantation, and the majority also reported improved muscle, endothelium, and/or nerve in the erectile tissue. However, differentiation or engraftment of transplanted SCs has rarely been observed; thus, paracrine action is generally believed to be responsible for SC’s therapeutic effects. But still, few studies actually investigated and none proved paracrine action as a therapeutic mechanism. Thus, based exclusively on functional outcome data shown in preclinical studies, two clinical trials are currently recruiting patients for treatment with IC injection of ADSC and BMSC, respectively. 1. Introduction Erectile dysfunction (ED) is a term recommended by a panel of experts in 1992 to replace the term “impotence” [1]. These experts also defined ED as an inability of the male to attain and/or maintain penile erection sufficient for satisfactory sexual performance. Although not life threatening by itself, ED is a strong predictor of high-mortality diseases such as coronary artery disease and cardiovascular disease [2–5]. ED does directly and negatively impact the quality of life of the afflicted men and their spouse [6–11]. In a 1999 report the worldwide prevalence of ED was estimated to be 152 million men in 1995 and predicted to increase to 322 million men by 2025 [12]. While the majority of ED cases can be treated with currently available medications or devices, approximately 20% of the overall ED patient population remains unresponsive to treatment [13], and in certain patient populations, such as those having diabetes mellitus (DM) or having undergone radical prostatectomy (RP), the failure rates are even higher, at 40% [14–16]. Moreover, regardless of their therapeutic efficacy or inefficacy, all current treatment options treat only the symptoms, not

References

[1]  N. C. Panel, “Impotence,” NIH Consens Statement, vol. 10, pp. 1–33, 1992.
[2]  B. A. Inman, J. L. St. Sauver, D. J. Jacobson et al., “A population-based, longitudinal study of erectile dysfunction and future coronary artery disease,” Mayo Clinic Proceedings, vol. 84, no. 2, pp. 109–113, 2009.
[3]  G. Jackson, N. Boon, I. Eardley et al., “Erectile dysfunction and coronary artery disease prediction: evidence-based guidance and consensus,” International Journal of Clinical Practice, vol. 64, no. 7, pp. 848–857, 2010.
[4]  K. Chew, J. Finn, B. Stuckey et al., “Erectile dysfunction as a predictor for subsequent atherosclerotic cardiovascular events: findings from a linked-data study,” The Journal of Sexual Medicine, vol. 7, no. 1, pp. 192–202, 2010.
[5]  J. Dong, Y. Zhang, and L. Qin, “Erectile dysfunction and risk of cardiovascular disease: meta-analysis of prospective cohort studies,” Journal of the American College of Cardiology, vol. 58, no. 13, pp. 1378–1385, 2011.
[6]  G. Wagner, K. S. Fugl-Meyer, and A. R. Fugl-Meyer, “Impact of erectile dysfunction on quality of life: patient and partner perspectives,” International Journal of Impotence Research, vol. 12, supplement 4, pp. S144–S146, 2000.
[7]  J. J. Sánchez-Cruz, A. Cabrera-León, A. Martín-Morales, A. Fernández, R. Burgos, and J. Rejas, “Male erectile dysfunction and health-related quality of life,” European Urology, vol. 44, no. 2, pp. 245–253, 2003.
[8]  T. Kushiro, A. Takahashi, F. Saito et al., “Erectile dysfunction and its influence on quality of life in patients with essential hypertension,” American Journal of Hypertension, vol. 18, no. 3, pp. 427–430, 2005.
[9]  M. S. Sand, W. Fisher, R. Rosen, J. Heiman, and I. Eardley, “Erectile dysfunction and constructs of masculinity and quality of life in the multinational Men's Attitudes to Life Events and Sexuality (MALES) study,” The Journal of Sexual Medicine, vol. 5, no. 3, pp. 583–594, 2008.
[10]  G. V. Fernandes, R. R. dos Santos, W. Soares et al., “The impact of erectile dysfunction on the quality of life of men undergoing hemodialysis and its association with depression,” The Journal of Sexual Medicine, vol. 7, no. 12, pp. 4003–4010, 2010.
[11]  A. Avasthi, S. Grover, A. Bhansali et al., “Erectile dysfunction in diabetes mellitus contributes to poor quality of life,” International Review of Psychiatry, vol. 23, no. 1, pp. 93–99, 2011.
[12]  I. A. Ayta?, J. B. McKinlay, and R. J. Krane, “The likely worldwide increase in erectile dysfunction between 1995 and 2025 and some possible policy consequences,” BJU International, vol. 84, no. 1, pp. 50–56, 1999.
[13]  A. Melman and K. Davies, “Gene therapy for erectile dysfunction: what is the future?” Current Urology Reports, vol. 11, no. 6, pp. 421–426, 2010.
[14]  A. R. McCullough, J. H. Barada, A. Fawzy, A. T. Guay, and D. Hatzichristou, “Achieving treatment optimization with sildenafil citrate (Viagra) in patients with erectile dysfunction,” Urology, vol. 60, no. 2, pp. 28–38, 2002.
[15]  M. Kendirci and W. J. G. Hellstrom, “Current concepts in the management of erectile dysfunction in men with prostate cancer,” Clinical Prostate Cancer, vol. 3, no. 2, pp. 87–92, 2004.
[16]  V. Phé and M. Rouprêt, “Erectile dysfunction and diabetes: a review of the current evidence-based medicine and a synthesis of the main available therapies,” Diabetes and Metabolism, vol. 38, no. 1, pp. 1–13, 2012.
[17]  T. F. Lue, “Erectile dysfunction,” The New England Journal of Medicine, vol. 342, no. 24, pp. 1802–1813, 2000.
[18]  G. Lin, X. Qiu, T. M. Fandel et al., “Improved penile histology by phalloidin stain: circular and longitudinal cavernous smooth muscles, dual-endothelium arteries, and erectile dysfunction-associated changes,” Urology, vol. 78, no. 4, pp. 970.e1–970.e8, 2011.
[19]  X. Qiu, T. M. Fandel, G. Lin et al., “Cavernous smooth muscle hyperplasia in a rat model of hyperlipidaemia- associated erectile dysfunction,” BJU International, vol. 108, no. 11, pp. 1866–1872, 2011.
[20]  A. Awad, B. Alsaid, T. Bessede, S. Droupy, and G. Beno?t, “Evolution in the concept of erection anatomy,” Surgical and Radiologic Anatomy, vol. 33, no. 4, pp. 301–312, 2011.
[21]  W. G. Dail, D. Trujillo, D. de la Rosa, and G. Walton, “Autonomic innervation of reproductive organs: analysis of the neurons whose axons project in the main penile nerve in the pelvic plexus of the rat,” Anatomical Record, vol. 224, no. 1, pp. 94–101, 1989.
[22]  C. S. Lin, “Lue TF Cyclic nucleotide signaling in vascular and cavernous smooth muscle: aging-related changes,” in Advances in Cell Aging and Gerontology, E. Bittar, Ed., vol. 16, pp. 57–106, Elsevier, 2004.
[23]  C. Lin, G. Lin, and T. F. Lue, “Cyclic nucleotide signaling in cavernous smooth muscle,” The Journal of Sexual Medicine, vol. 2, no. 4, pp. 478–491, 2005.
[24]  C. Lin, Z. Xin, Z. Wang, G. Lin, and T. F. Lue, “Molecular Yin and Yang of erectile function and dysfunction,” Asian Journal of Andrology, vol. 10, no. 3, pp. 433–440, 2008.
[25]  K. J. Hurt, B. Musicki, M. A. Palese et al., “Akt-dependent phosphorylation of endothelial nitric-oxide synthase mediates penile erection,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 6, pp. 4061–4066, 2002.
[26]  T. J. Bivalacqua, M. F. Usta, H. C. Champion, P. J. Kadowitz, and W. J. G. Hellstrom, “Endothelial dysfunction in erectile dysfunction: role of the endothelium in erectile physiology and disease,” Journal of Andrology, vol. 24, pp. S17–S37, 2003.
[27]  A. L. Burnett, “Novel nitric oxide signaling mechanisms regulate the erectile response,” International Journal of Impotence Research, vol. 16, supplement 1, pp. S15–S19, 2004.
[28]  G. Corona, L. Petrone, E. Mannucci et al., “Difficulties in achieving vs maintaining erection: organic, psychogenic and relational determinants,” International Journal of Impotence Research, vol. 17, no. 3, pp. 252–258, 2005.
[29]  H. Wessells, T. H. Teal, K. Engel et al., “Fluid shear stress-induced nitric oxide production in human cavernosal endothelial cells: inhibition by hyperglycaemia,” BJU International, vol. 97, no. 5, pp. 1047–1052, 2006.
[30]  T. Michel and O. Feron, “Nitric oxide synthases: which, where, how, and why?” The Journal of Clinical Investigation, vol. 100, no. 9, pp. 2146–2152, 1997.
[31]  C. Lin, A. Lau, E. Bakircioglu et al., “Analysis of neuronal nitric oxide synthase isoform expression and identification of human nNOS-μ,” Biochemical and Biophysical Research Communications, vol. 253, no. 2, pp. 388–394, 1998.
[32]  H. Ning, X. Qiu, L. Baine, et al., “Effects of high glucose on human cavernous endothelial cells,” Urology, vol. 80, pp. 1162. e7–1162. e11, 2012.
[33]  C. Lin, “Phosphodiesterase type 5 regulation in the penile corpora cavernosa,” The Journal of Sexual Medicine, vol. 6, supplement 3, pp. 203–209, 2009.
[34]  C. Lin, G. Lin, Z. Xin, and T. F. Lue, “Expression, distribution and regulation of phosphodiesterase 5,” Current Pharmaceutical Design, vol. 12, no. 27, pp. 3439–3457, 2006.
[35]  R. Shamloul and H. Ghanem, “Erectile dysfunction,” The Lancet, vol. 381, pp. 153–165, 2013.
[36]  A. A. Carvalheira, N. M. Pereira, J. Maroco, et al., “Dropout in the treatment of erectile dysfunction with PDE5: a study on predictors and a qualitative analysis of reasons for discontinuation,” The Journal of Sexual Medicine, vol. 9, pp. 2361–2369, 2012.
[37]  National Diabetes Statistics, National Diabetes Information Clearinghouse, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, 2011, http://diabetes.niddk.nih.gov/dm/pubs/statistics/index.aspx.
[38]  G. Danaei, M. M. Finucane, Y. Lu et al., “National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2·7 million participants,” The Lancet, vol. 378, no. 9785, pp. 31–40, 2011.
[39]  IDF Diabetes Atlas, International Diabetes Federation, http://www.idf.org/diabetesatlas/5e/the-global-burden.
[40]  R. W. Lewis, K. S. Fugl-Meyer, G. Corona et al., “Definitions/epidemiology/risk factors for sexual dysfunction,” The Journal of Sexual Medicine, vol. 7, no. 4, pp. 1598–1607, 2010.
[41]  H. A. Feldman, I. Goldstein, D. G. Hatzichristou, R. J. Krane, and J. B. McKinlay, “Impotence and its medical and psychosocial correlates: results of the Massachusetts Male Aging study,” Journal of Urology, vol. 151, no. 1, pp. 54–61, 1994.
[42]  A. Ponholzer, C. Temml, K. Mock, M. Marszalek, R. Obermayr, and S. Madersbacher, “Prevalence and risk factors for erectile dysfunction in 2869 men using a validated questionnaire,” European Urology, vol. 47, no. 1, pp. 80–85, 2005.
[43]  V. Fonseca, A. Seftel, J. Denne, and P. Fredlund, “Impact of diabetes mellitus on the severity of erectile dysfunction and response to treatment: analysis of data from tadalafil clinical trials,” Diabetologia, vol. 47, no. 11, pp. 1914–1923, 2004.
[44]  D. F. Penson, D. M. Latini, D. P. Lubeck, K. L. Wallace, J. M. Henning, and T. F. Lue, “Do impotent men with diabetes have more severe erectile dysfunction and worse quality of life than the general population of impotent patients? Results from the Exploratory Comprehensive Evaluation of Erectile Dysfunction (ExCEED) database,” Diabetes Care, vol. 26, no. 4, pp. 1093–1099, 2003.
[45]  M. Albersen, G. Lin, T. M. Fandel et al., “Functional, metabolic, and morphologic characteristics of a novel rat model of type 2 diabetes-associated erectile dysfunction,” Urology, vol. 78, no. 2, pp. 476.e1–476.e8, 2011.
[46]  M. R. Dashwood, A. Crump, X. Shi-Wen, and A. Loesch, “Identification of neuronal nitric oxide synthase (nNOS) in human penis: a potential role of reduced neuronally-derived nitric oxide in erectile dysfunction,” Current Pharmaceutical Biotechnology, vol. 12, no. 9, pp. 1316–1321, 2011.
[47]  F. Zhou, H. Xin, T. Liu, et al., “Effects of icariside II on improving erectile function in rats with streptozotocin-induced diabetes,” Journal of Andrology, vol. 33, pp. 832–844, 2012.
[48]  S. Cellek, N. A. Foxwell, and S. Moncada, “Two phases of nitrergic neuropathy in streptozotocin-induced diabetic rats,” Diabetes, vol. 52, no. 9, pp. 2353–2362, 2003.
[49]  X. Qiu, G. Lin, Z. Xin, et al., “Effects of low-energy shockwave therapy on the erectile function and tissue of a diabetic rat model,” The Journal of Sexual Medicine, vol. 10, pp. 738–746, 2013.
[50]  C. Costa, R. Soares, ?. Castela et al., “Increased endothelial apoptotic cell density in human diabetic erectile tissue—comparison with clinical data,” The Journal of Sexual Medicine, vol. 6, no. 3, pp. 826–835, 2009.
[51]  H. Ning, G. Lin, T. F. Lue, et al., “A coculture system of cavernous endothelial and smooth muscle cells,” International Journal of Impotence Research, vol. 25, pp. 63–68, 2013.
[52]  K. Park, K. S. Ryu, W. J. Li, S. W. Kim, and J. Paick, “Chronic treatment with a type 5 phosphodiesterase inhibitor suppresses apoptosis of corporal smooth muscle by potentiating Akt signalling in a rat model of diabetic erectile dysfunction,” European Urology, vol. 53, no. 6, pp. 1282–1288, 2008.
[53]  A. Jemal, R. Siegel, J. Xu, and E. Ward, “Cancer statistics,” CA Cancer Journal for Clinicians, vol. 60, no. 5, pp. 277–300, 2010.
[54]  J. Brandeis, C. L. Pashos, J. M. Henning, et al., “A nationwide charge comparison of the principal treatments for early stage prostate carcinoma,” Cancer, vol. 89, pp. 1792–1799, 2000.
[55]  R. C. Dean and T. F. Lue, “Neuroregenerative strategies after radical prostatectomy,” Reviews in Urology, vol. 7, supplement 2, pp. S26–S32, 2005.
[56]  M. Kendirci, J. Bejma, and W. J. G. Hellstrom, “Update on erectile dysfunction in prostate cancer patients,” Current Opinion in Urology, vol. 16, no. 3, pp. 186–195, 2006.
[57]  M. J. Resnick, T. Koyama, K. H. Fan, et al., “Long-term functional outcomes after treatment for localized prostate cancer,” The New England Journal of Medicine, vol. 368, pp. 436–445, 2013.
[58]  V. Ficarra, G. Novara, T. E. Ahlering, et al., “Systematic review and meta-analysis of studies reporting potency rates after robot-assisted radical prostatectomy,” European Urology, vol. 62, pp. 418–430, 2012.
[59]  Y. Pardo, F. Guedea, F. Aguilo, et al., “Quality-of-life impact of primary treatments for localized prostate cancer in patients without hormonal treatment,” Journal of Clinical Oncology, vol. 28, pp. 4687–4696, 2010.
[60]  M. Fode, D. A. Ohl, D. Ralph, et al., “Penile rehabilitation after radical prostatectomy: what the evidence really says,” BJU International, vol. 112, no. 7, pp. 998–1008, 2013.
[61]  M. Albersen, M. Kendirci, F. van der Aa, W. J. G. Hellstrom, T. F. Lue, and J. L. Spees, “Multipotent stromal cell therapy for cavernous nerve injury-induced erectile dysfunction,” The Journal of Sexual Medicine, vol. 9, no. 2, pp. 385–403, 2012.
[62]  M. G. Ferrini, H. H. Davila, I. Kovanecz, S. P. Sanchez, N. F. Gonzalez-Cadavid, and J. Rajfer, “Vardenafil prevents fibrosis and loss of corporal smooth muscle that occurs after bilateral cavernosal nerve resection in the rat,” Urology, vol. 68, no. 2, pp. 429–435, 2006.
[63]  L. T. Klein, M. I. Miller, R. Buttyan et al., “Apoptosis in the rat penis after penile denervation,” Journal of Urology, vol. 158, no. 2, pp. 626–630, 1997.
[64]  J. J. Lysiak, S. Yang, A. P. Klausner, H. Son, J. B. Tuttle, and W. D. Steers, “Tadalafil increases Akt and extracellular signal-regulated kinase 1/2 activation, and prevents apoptotic cell death in the penis following denervation,” Journal of Urology, vol. 179, no. 2, pp. 779–785, 2008.
[65]  S. Leungwattanakij, T. J. Bivalacqua, M. F. Usta et al., “Cavernous neurotomy causes hypoxia and fibrosis in rat corpus cavernosum,” Journal of Andrology, vol. 24, no. 2, pp. 239–245, 2003.
[66]  W. Hu, L. Hu, J. Song et al., “Fibrosis of corpus cavernosum in animals following cavernous nerve ablation,” Asian Journal of Andrology, vol. 6, no. 2, pp. 111–116, 2004.
[67]  F. Iacono, R. Giannella, P. Somma, G. Manno, F. Fusco, and V. Mirone, “Histological alterations in cavernous tissue after radical prostatectomy,” Journal of Urology, vol. 173, no. 5, pp. 1673–1676, 2005.
[68]  S. Carrier, H. Hricak, S.-S. Lee et al., “Radiation-induced decrease in nitric oxide synthase—containing nerves in the rat penis,” Radiology, vol. 195, no. 1, pp. 95–99, 1995.
[69]  M. Kimura, H. Yan, Z. Rabbani et al., “Radiation-induced erectile dysfunction using prostate-confined modern radiotherapy in a rat model,” The Journal of Sexual Medicine, vol. 8, no. 8, pp. 2215–2226, 2011.
[70]  G. Garaffa, L. W. Trost, E. C. Serefoglu, et al., “Understanding the course of Peyronie's disease,” International Journal of Clinical Practice, vol. 67, pp. 781–788, 2013.
[71]  T. J. Walsh, J. M. Hotaling, T. F. Lue, et al., “How curved is too curved? The severity of penile deformity may predict sexual disability among men with Peyronie's disease,” International Journal of Impotence Research, vol. 25, pp. 109–112, 2013.
[72]  J. A. Lopez and J. P. Jarow, “Penile vascular evaluation of men with Peyronie's disease,” Journal of Urology, vol. 149, no. 1, pp. 53–55, 1993.
[73]  M. ?ulha, B. Alici, O. Acar, N. Mutlu, and A. G?kalp, “The relationship between diabetes mellitus, impotence and veno-occlusive dysfunction in Peyronie's disease patients,” Urologia Internationalis, vol. 60, no. 2, pp. 101–104, 1998.
[74]  J. F. Stecker Jr. and C. J. Devine Jr., “Evaluation of erectile dysfunction in patients with Peyronie's disease,” Journal of Urology, vol. 132, no. 4, pp. 680–681, 1984.
[75]  T. J. Bivalacqua, E. K. Diner, T. E. Novak et al., “A rat model of Peyronie's disease associated with a decrease in erectile activity and an increase in inducible nitric oxide synthase protein expression,” Journal of Urology, vol. 163, no. 6, pp. 1992–1998, 2000.
[76]  D. Martinez, C. E. Ercole, T. S. Hakky, et al., “Peyronie's disease: still a surgical disease,” Advances in Urology, vol. 2012, Article ID 206284, 5 pages, 2012.
[77]  A. Kadioglu, O. Sanli, T. Akman, et al., “Graft materials in Peyronie's disease surgery: a comprehensive review,” The Journal of Sexual Medicine, vol. 4, pp. 581–595, 2007.
[78]  L. Ma, Y. Yang, S. C. Sikka et al., “Adipose tissue-derived stem cell-seeded small intestinal submucosa for tunica albuginea grafting and reconstruction,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 6, pp. 2090–2095, 2012.
[79]  F. Castiglione, P. Hedlund, F. van der Aa, et al., “Intratunical injection of human adipose tissue-derived stem cells prevents fibrosis and is associated with improved erectile function in a rat model of Peyronie's disease,” European Urology, vol. 63, pp. 551–560, 2013.
[80]  J. Y. Bahk, J. H. Jung, H. Han, S. K. Min, and Y. S. Lee, “Treatment of diabetic impotence with umbilical cord blood stem cell intracavernosal transplant: preliminary report of 7 cases,” Experimental and Clinical Transplantation, vol. 8, no. 2, pp. 150–160, 2010.
[81]  T. J. Bivalacqua, W. Deng, M. Kendirci et al., “Mesenchymal stem cells alone or ex vivo gene modified with endothelial nitric oxide synthase reverse age-associated erectile dysfunction,” American Journal of Physiology—Heart and Circulatory Physiology, vol. 292, no. 3, pp. H1278–H1290, 2007.
[82]  G. Nolazco, I. Kovanecz, D. Vernet et al., “Effect of muscle-derived stem cells on the restoration of corpora cavernosa smooth muscle and erectile function in the aged rat,” BJU International, vol. 101, no. 9, pp. 1156–1164, 2008.
[83]  M. T. Abdel Aziz, S. El-Haggar, T. Mostafa et al., “Effect of mesenchymal stem cell penile transplantation on erectile signaling of aged rats,” Andrologia, vol. 42, no. 3, pp. 187–192, 2010.
[84]  Y. Huang, H. Ning, A. W. Shindel et al., “The effect of intracavernous injection of adipose tissue-derived stem cells on hyperlipidemia-associated erectile dysfunction in a rat model,” The Journal of Sexual Medicine, vol. 7, no. 4, pp. 1391–1400, 2010.
[85]  M. M. Garcia, T. M. Fandel, G. Lin et al., “Treatment of erectile dysfunction in the obese Type 2 diabetic ZDF rat with adipose tissue-derived stem cells,” The Journal of Sexual Medicine, vol. 7, no. 1, pp. 89–98, 2010.
[86]  X. Gou, W. He, M. Xiao et al., “Transplantation of endothelial progenitor cells transfected with VEGF165 to restore erectile function in diabetic rats,” Asian Journal of Andrology, vol. 13, no. 2, pp. 332–338, 2011.
[87]  X. Qiu, H. Lin, Y. Wang et al., “Intracavernous transplantation of bone marrow-derived mesenchymal stem cells restores erectile function of streptozocin-induced diabetic rats,” The Journal of Sexual Medicine, vol. 8, no. 2, pp. 427–436, 2011.
[88]  X. Qiu, C. Sun, W. Yu et al., “Combined strategy of mesenchymal stem cell injection with vascular endothelial growth factor gene therapy for the treatment of diabetes-associated erectile dysfunction,” Journal of Andrology, vol. 33, no. 1, pp. 37–44, 2012.
[89]  C. Sun, H. Lin, W. Yu et al., “Neurotrophic effect of bone marrow mesenchymal stem cells for erectile dysfunction in diabetic rats,” International Journal of Andrology, vol. 35, pp. 601–607, 2012.
[90]  H. Nishimatsu, E. Suzuki, S. Kumano et al., “Adrenomedullin mediates adipose tissue-derived stem cell-induced restoration of erectile function in diabetic rats,” The Journal of Sexual Medicine, vol. 9, no. 2, pp. 482–493, 2012.
[91]  J. K. Ryu, M. Tumurbaatar, H. R. Jin, et al., “Intracavernous delivery of freshly isolated stromal vascular fraction rescues erectile function by enhancing endothelial regeneration in the streptozotocin-induced diabetic mouse,” The Journal of Sexual Medicine, vol. 9, pp. 3051–3065, 2012.
[92]  Y. He, W. He, G. Qin, et al., “Transplantation KCNMA1 modified bone marrow-mesenchymal stem cell therapy for diabetes mellitus-induced erectile dysfunction,” Andrologia, 2013.
[93]  G. Liu, X. Sun, J. Bian, et al., “Correction of diabetic erectile dysfunction with adipose derived stem cells modified with the vascular endothelial growth factor gene in a rodent diabetic model,” PLoS ONE, vol. 8, Article ID e72790, 2013.
[94]  D. Bochinski, G. T. Lin, L. Nunes et al., “The effect of neural embryonic stem cell therapy in a rat model of cavernosal nerve injury,” BJU International, vol. 94, no. 6, pp. 904–909, 2004.
[95]  Y. Kim, F. de Miguel, I. Usiene et al., “Injection of skeletal muscle-derived cells into the penis improves erectile function,” International Journal of Impotence Research, vol. 18, no. 4, pp. 329–334, 2006.
[96]  P. A. Fall, M. Izikki, L. Tu, et al., “Apoptosis and effects of intracavernous bone marrow cell injection in a rat model of postprostatectomy erectile dysfunction,” European Urology, vol. 56, pp. 716–725, 2009.
[97]  M. Albersen, T. M. Fandel, G. Lin et al., “Injections of adipose tissue-derived stem cells and stem cell lysate improve recovery of erectile function in a rat model of cavernous nerve injury,” The Journal of Sexual Medicine, vol. 7, no. 10, pp. 3331–3340, 2010.
[98]  M. Kendirci, L. Trost, B. Bakondi, M. J. Whitney, W. J. G. Hellstrom, and J. L. Spees, “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,” Journal of Urology, vol. 184, no. 4, pp. 1560–1566, 2010.
[99]  G. Lin, M. Albersen, A. M. Harraz et al., “Cavernous nerve repair with allogenic adipose matrix and autologous adipose-derived stem cells,” Urology, vol. 77, no. 6, pp. 1509.e1–1509.e8, 2011.
[100]  J. C. Woo, W. J. Bae, S. J. Kim et al., “Transplantation of muscle-derived stem cells into the corpus avernosum restores erectile function in a rat model of cavernous nerve injury,” Korean Journal of Urology, vol. 52, no. 5, pp. 359–363, 2011.
[101]  T. M. Fandel, M. Albersen, G. Lin 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,” European Urology, vol. 61, no. 1, pp. 201–210, 2012.
[102]  S. J. Kim, S. W. Choi, K. J. Hur, et al., “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 Journal of Urology, vol. 53, pp. 726–732, 2012.
[103]  I. Kovanecz, S. Rivera, G. Nolazco, et al., “Separate or combined treatments with daily sildenafil, molsidomine, or muscle-derived stem cells prevent erectile dysfunction in a rat model of cavernosal nerve damage,” The Journal of Sexual Medicine, vol. 9, pp. 2814–2826, 2012.
[104]  S. Piao, I. G. Kim, J. Y. Lee, et al., “Therapeutic effect of adipose-derived stem cells and BDNF-immobilized PLGA membrane in a rat model of cavernous nerve injury,” The Journal of Sexual Medicine, vol. 9, pp. 1968–1979, 2012.
[105]  X. Qiu, J. Villalta, L. Ferretti et al., “Effects of intravenous injection of adipose-derived stem cells in a rat model of radiation therapy-induced erectile dysfunction,” The Journal of Sexual Medicine, vol. 9, pp. 1834–1841, 2012.
[106]  X. Qiu, T. M. Fandel, L. Ferretti, et al., “Both immediate and delayed intracavernous injection of autologous adipose-derived stromal vascular fraction enhances recovery of erectile function in a rat model of cavernous nerve injury,” European Urology, vol. 62, pp. 720–727, 2012.
[107]  H. H. Jeong, S. Piao, J. N. Ha, et al., “Combined therapeutic effect of udenafil and adipose-derived stem cell (ADSC)/brain-derived neurotrophic factor (BDNF)-membrane system in a rat model of cavernous nerve injury,” Urology, vol. 81, pp. 1108. e7–1108. e14, 2013.
[108]  I. G. Kim, S. Piao, J. Y. Lee, et al., “Effect of an adipose-derived stem cell and nerve growth factor-incorporated hydrogel on recovery of erectile function in a rat model of cavernous nerve injury,” Tissue Engineering A, vol. 19, pp. 14–23, 2013.
[109]  D. You, M. J. Jang, J. Lee, 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, vol. 81, pp. 104–110, 2013.
[110]  D. You, M. J. Jang, J. Lee, 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, vol. 73, pp. 278–286, 2013.
[111]  W. Y. Choi, H. G. Jeon, Y. Chung, et al., “Isolation and characterization of novel, highly proliferative human CD34/CD73-double-positive testis-derived stem cells for cell therapy,” Stem Cells and Development, vol. 22, pp. 2158–2173, 2013.
[112]  C. Ying, M. Yang, X. Zheng, et al., “Effects of intracavernous injection of adipose-derived stem cells on cavernous nerve regeneration in a rat model,” Cellular and Molecular Neurobiology, vol. 33, pp. 233–240, 2013.
[113]  B. F. Koontz, H. Yan, M. Kimura, Z. Vujaskovic, C. Donatucci, and F. Yin, “Feasibility study of an intensity-modulated radiation model for the study of erectile dysfunction,” The Journal of Sexual Medicine, vol. 8, no. 2, pp. 411–418, 2011.
[114]  G. J. van der Wielen, M. Vermeij, B. W. D. de Jong et al., “Changes in the penile arteries of the rat after fractionated irradiation of the prostate: a pilot study,” The Journal of Sexual Medicine, vol. 6, no. 7, pp. 1908–1913, 2009.
[115]  L. da Silva Meirelles, P. C. Chagastelles, and N. B. Nardi, “Mesenchymal stem cells reside in virtually all post-natal organs and tissues,” Journal of Cell Science, vol. 119, no. 11, pp. 2204–2213, 2006.
[116]  M. Crisan, S. Yap, L. Casteilla et al., “A perivascular origin for mesenchymal stem cells in multiple human organs,” Cell Stem Cell, vol. 3, no. 3, pp. 301–313, 2008.
[117]  G. Lin, M. Garcia, H. Ning et al., “Defining stem and progenitor cells within adipose tissue,” Stem Cells and Development, vol. 17, no. 6, pp. 1053–1063, 2008.
[118]  J. Braun, A. Kurtz, N. Barutcu, et al., “Concerted regulation of CD34 and CD105 accompanies mesenchymal stromal cell derivation from human adventitial stromal cell,” Stem Cells and Development, vol. 22, pp. 815–827, 2013.
[119]  M. Corselli, C. W. Chen, B. Sun, et al., “The tunica adventitia of human arteries and veins as a source of mesenchymal stem cells,” Stem Cells and Development, vol. 21, pp. 1299–1308, 2012.
[120]  G. Lin, Z. Xin, H. Zhang et al., “Identification of active and quiescent adipose vascular stromal cells,” Cytotherapy, vol. 14, no. 2, pp. 240–246, 2012.
[121]  M. Maumus, J.-A. Peyrafitte, R. D'Angelo et al., “Native human adipose stromal cells: localization, morphology and phenotype,” International Journal of Obesity, vol. 35, no. 9, pp. 1141–1153, 2011.
[122]  L. Zimmerlin, V. S. Donnenberg, M. E. Pfeifer et al., “Stromal vascular progenitors in adult human adipose tissue,” Cytometry A, vol. 77, no. 1, pp. 22–30, 2010.
[123]  C. S. Lin and T. F. Lue, “Defining vascular stem cells,” Stem Cells and Development, vol. 22, pp. 1018–1026, 2013.
[124]  M. Dominici, K. Le Blanc, I. Mueller et al., “Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement,” Cytotherapy, vol. 8, no. 4, pp. 315–317, 2006.
[125]  C. S. Lin and T. F. Lue, “Adipose-derived stem cells: therapy through paracrine actions,” in Stem Cells and Cancer Stem Cells, M. A. Hayat, Ed., vol. 4, pp. 203–216, Springer, New York, NY, USA, 2012.
[126]  C. S. Lin, Z. C. Xin, J. Dai, et al., “Commonly used mesenchymal stem cell markers and tracking labels: limitations and challenges,” Histology and Histopathology, vol. 28, pp. 1109–1116, 2013.
[127]  C. S. Lin, H. Ning, G. Lin, et al., “Is CD34 truly a negative marker for mesenchymal stromal cells?” Cytotherapy, vol. 14, pp. 1159–1163, 2012.
[128]  P. J. Simmons and B. Torok-Storb, “Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1,” Blood, vol. 78, no. 1, pp. 55–62, 1991.
[129]  P. J. Simmons and B. Torok-Storb, “CD34 expression by stromal precursors in normal human adult bone marrow,” Blood, vol. 78, no. 11, pp. 2848–2853, 1991.
[130]  C. M. Kolf, E. Cho, and R. S. Tuan, “Mesenchymal stromal cells. Biology of adult mesenchymal stem cells: regulation of niche, self-renewal and differentiation,” Arthritis Research and Therapy, vol. 9, no. 1, pp. 204–213, 2007.
[131]  G. Lin, G. Liu, L. Banie et al., “Tissue distribution of mesenchymal stem cell marker stro-1,” Stem Cells and Development, vol. 20, no. 10, pp. 1747–1752, 2011.
[132]  A. I. Caplan and D. Correa, “The MSC: an injury drugstore,” Cell Stem Cell, vol. 9, no. 1, pp. 11–15, 2011.
[133]  D. J. Prockop and J. Y. Oh, “Medical therapies with adult stem/progenitor cells (MSCs): a backward journey from dramatic results in vivo to the cellular and molecular explanations,” Journal of Cellular Biochemistry, vol. 113, no. 5, pp. 1460–1469, 2012.
[134]  M. B. Murphy, K. Moncivais, and A. I. Caplan, “Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine,” Experimental & Molecular Medicine, vol. 45, article e54, 2013.
[135]  D. Brenin, J. Look, M. Bader, N. Hübner, G. Levan, and P. Iannaccone, “Rat embryonic stem cells: a progress report,” Transplantation Proceedings, vol. 29, no. 3, pp. 1761–1765, 1997.
[136]  M. Kawamata and T. Ochiya, “Establishment of embryonic stem cells from rat blastocysts,” Methods in Molecular Biology, vol. 597, pp. 169–177, 2010.
[137]  M. Schulze, H. Ungefroren, M. Bader, and F. F?ndrich, “Derivation, maintenance, and characterization of rat embryonic stem cells in vitro,” Methods in Molecular Biology, vol. 329, pp. 45–58, 2006.
[138]  H. Ning, G. Lin, T. F. Lue, and C. Lin, “Neuron-like differentiation of adipose tissue-derived stromal cells and vascular smooth muscle cells,” Differentiation, vol. 74, no. 9-10, pp. 510–518, 2006.
[139]  A. B. Mathiasen, E. Jorgensen, A. A. Qayyum, et al., “Rationale and design of the first randomized, double-blind, placebo-controlled trial of intramyocardial injection of autologous bone-marrow derived Mesenchymal Stromal Cells in chronic ischemic Heart Failure (MSC-HF Trial),” American Heart Journal, vol. 164, pp. 285–291, 2012.
[140]  T. Asahara and J. M. Isner, “Endothelial progenitor cells for vascular regeneration,” Journal of Hematotherapy and Stem Cell Research, vol. 11, no. 2, pp. 171–178, 2002.
[141]  C. Foresta, N. Caretta, A. Lana, A. Cabrelle, G. Palù, and A. Ferlin, “Circulating endothelial progenitor cells in subjects with erectile dysfunction,” International Journal of Impotence Research, vol. 17, no. 3, pp. 288–290, 2005.
[142]  C. Foresta, N. Caretta, A. Lana et al., “Relationship between vascular damage degrees and endothelial progenitor cells in patients with erectile dysfunction: effect of vardenafil administration and PDE5 expression in the bone marrow,” European Urology, vol. 51, no. 5, pp. 1411–1419, 2007.
[143]  C. Foresta, A. Ferlin, L. de Toni et al., “Circulating endothelial progenitor cells and endothelial function after chronic Tadalafil treatment in subjects with erectile dysfunction,” International Journal of Impotence Research, vol. 18, no. 5, pp. 484–488, 2006.
[144]  M. Baumh?kel, N. Werner, M. B?hm, and G. Nickenig, “Circulating endothelial progenitor cells correlate with erectile function in patients with coronary heart disease,” European Heart Journal, vol. 27, no. 18, pp. 2184–2188, 2006.
[145]  K. Esposito, M. Ciotola, M. I. Maiorino et al., “Circulating CD34+KDR+ endothelial progenitor cells correlate with erectile function and endothelial function in overweight men,” The Journal of Sexual Medicine, vol. 6, no. 1, pp. 107–114, 2009.
[146]  T. E. Ichim, Z. Zhong, N. A. Mikirova et al., “Circulating endothelial progenitor cells and erectile dysfunction: possibility of nutritional intervention?” Panminerva Medica, vol. 52, no. 2, pp. 75–80, 2010.
[147]  S. la Vignera, R. Condorelli, E. Vicari, R. D'Agata, and A. E. Calogero, “Circulating endothelial progenitor cells and endothelial microparticles in patients with arterial erectile dysfunction and metabolic syndrome,” Journal of Andrology, vol. 33, no. 2, pp. 202–209, 2012.
[148]  S. la Vignera, R. Condorelli, E. Vicari, R. D'Agata, and A. Calogero, “Original immunophenotype of blood endothelial progenitor cells and microparticles in patients with isolated arterial erectile dysfunction and late onset hypogonadism: effects of androgen replacement therapy,” Aging Male, vol. 14, no. 3, pp. 183–189, 2011.
[149]  S. la Vignera, R. A. Condorelli, E. Vicari, R. D'Agata, and A. E. Calogero, “New immunophenotype of blood endothelial progenitor cells and endothelial microparticles in patients with arterial erectile dysfunction and late-onset hypogonadism,” Journal of Andrology, vol. 32, no. 5, pp. 509–517, 2011.
[150]  M. C. Yoder and D. A. Ingram, “Endothelial progenitor cell: ongoing controversy for defining these cells and their role in neoangiogenesis in the murine system,” Current Opinion in Hematology, vol. 16, no. 4, pp. 269–273, 2009.
[151]  A. Mohammad and A. Shuaib, “Endothelial progenitor cells and vascular disease: are they for real?” Neurology, vol. 75, no. 23, pp. 2050–2051, 2010.
[152]  M. Prokopi, G. Pula, U. Mayr et al., “Proteomic analysis reveals presence of platelet microparticles in endothelial progenitor cell cultures,” Blood, vol. 114, no. 3, pp. 723–732, 2009.
[153]  M. C. Yoder, “Is endothelium the origin of endothelial progenitor cells?” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 30, pp. 1094–1103, 2010.
[154]  M. R. Richardson and M. C. Yoder, “Endothelial progenitor cells: quo vadis?” Journal of Molecular and Cellular Cardiology, vol. 50, no. 2, pp. 266–272, 2011.
[155]  M. C. Yoder, “Endothelial progenitor cell: a blood cell by many other names may serve similar functions,” Journal of Molecular Medicine, vol. 91, pp. 285–295, 2013.
[156]  F. Timmermans, J. Plum, M. C. Y?der, D. A. Ingram, B. Vandekerckhove, and J. Case, “Endothelial progenitor cells: identity defined?” Journal of Cellular and Molecular Medicine, vol. 13, no. 1, pp. 87–102, 2009.
[157]  J. Z. Shi, H. Zhang, M. Hou et al., “Is it possible to obtain “true endothelial progenitor cells” by in vitro culture of bone marrow mononuclear cells?” Stem Cells and Development, vol. 16, no. 4, pp. 683–690, 2007.
[158]  J. L. Goldberg, M. J. Laughlin, and V. J. Pompili, “Umbilical cord blood stem cells: implications for cardiovascular regenerative medicine,” Journal of Molecular and Cellular Cardiology, vol. 42, no. 5, pp. 912–920, 2007.
[159]  J. H. Park, I. Hwang, S. H. Hwang, et al., “Human umbilical cord blood-derived mesenchymal stem cells prevent diabetic renal injury through paracrine action,” Diabetes Research and Clinical Practice, vol. 98, pp. 465–473, 2012.
[160]  T. Yokoyama, R. Pruchnic, J. Y. Lee et al., “Autologous primary muscle-derived cells transfer into the lower urinary tract,” Tissue Engineering, vol. 7, no. 4, pp. 395–404, 2001.
[161]  T. Yokoyama, N. Yoshimura, R. Dhir et al., “Persistence and survival of autologous muscle derived cells versus bovine collagen as potential treatment of stress urinary incontinence,” Journal of Urology, vol. 165, no. 1, pp. 271–276, 2001.
[162]  A. Furuta, L. K. Carr, N. Yoshimura, et al., “Advances in the understanding of sress urinary incontinence and the promise of stem-cell therapy,” Reviews in Urology, vol. 9, pp. 106–112, 2007.
[163]  P. A. Zuk, M. Zhu, H. Mizuno et al., “Multilineage cells from human adipose tissue: implications for cell-based therapies,” Tissue Engineering, vol. 7, no. 2, pp. 211–228, 2001.
[164]  Y. C. Halvorsen, W. O. Wilkison, and J. M. Gimble, “Adipose-derived stromal cells—their utility and petential in bone formation,” International Journal of Obesity, vol. 24, supplement 4, pp. S41–S44, 2000.
[165]  S. Gronthos, D. M. Franklin, H. A. Leddy, et al., “Surface protein characterization of human adipose tissue-derived stromal cells,” Journal of Cellular Physiology, vol. 189, pp. 54–63, 2001.
[166]  Y. C. Halvorsen, A. Bond, A. Sen et al., “Thiazolidinediones and glucocorticoids synergistically induce differentiation of human adipose tissue stromal cells: biochemical, cellular, and molecular analysis,” Metabolism, vol. 50, no. 4, pp. 407–413, 2001.
[167]  Y.-D. C. Halvorsen, D. Franklin, A. L. Bond et al., “Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells,” Tissue Engineering, vol. 7, no. 6, pp. 729–741, 2001.
[168]  A. Sen, Y. R. Lea-Currie, D. Sujkowska, et al., “Adipogenic potential of human adipose derived stromal cells from multiple donors is heterogeneous,” Journal of Cellular Biochemistry, vol. 81, pp. 312–319, 2001.
[169]  P. Wu, K. Sato, S. Yukawa, Y. Hikasa, and K. Kagota, “Differentiation of stromal-vascular cells isolated from canine adipose tissues in primary culture,” Journal of Veterinary Medical Science, vol. 63, no. 1, pp. 17–23, 2001.
[170]  P. Pettersson, M. Cigolini, and L. Sjostrom, “Cells in human adipose tissue developing into adipocytes,” Acta Medica Scandinavica, vol. 215, no. 5, pp. 447–451, 1984.
[171]  B. M. Strem, K. C. Hicok, M. Zhu et al., “Multipotential differentiation of adipose tissue-derived stem cells,” Keio Journal of Medicine, vol. 54, no. 3, pp. 132–141, 2005.
[172]  T. K. Kim and J. H. Eberwine, “Mammalian cell transfection: the present and the future,” Analytical and Bioanalytical Chemistry, vol. 397, no. 8, pp. 3173–3178, 2010.
[173]  C. S. Lin, G. Lin, and T. F. Lue, “Allogeneic and xenogeneic transplantation of adipose-derived stem cells in immunocompetent recipients without immunosuppressants,” Stem Cells and Development, vol. 21, pp. 2770–2778, 2012.
[174]  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,” Journal of the American Medical Association, vol. 308, pp. 2369–2379, 2012.
[175]  G. Lin, X. Qiu, T. Fandel et al., “Tracking intracavernously injected adipose-derived stem cells to bone marrow,” International Journal of Impotence Research, vol. 23, no. 6, pp. 268–275, 2011.
[176]  H. Zhang, H. Ning, L. Banie et al., “Adipose tissue-derived stem cells secrete CXCL5 cytokine with chemoattractant and angiogenic properties,” Biochemical and Biophysical Research Communications, vol. 402, no. 3, pp. 560–564, 2010.
[177]  H. Zhang, R. Yang, Z. Wang, G. Lin, T. F. Lue, and C. Lin, “Adipose tissue-derived stem cells secrete CXCL5 cytokine with neurotrophic effects on cavernous nerve regeneration,” The Journal of Sexual Medicine, vol. 8, no. 2, pp. 437–446, 2011.
[178]  E. D. Kim, R. Nath, K. M. Slawin, D. Kadmon, B. J. Miles, and P. T. Scardino, “Bilateral nerve grafting during radical retropubic prostatectomy: extended follow-up,” Urology, vol. 58, no. 6, pp. 983–987, 2001.
[179]  B. A. Nelson, S. S. Chang, M. S. Cookson, and J. A. Smith Jr., “Morbidity and efficacy of genitofemoral nerve grafts with radical retropubic prostatectomy,” Urology, vol. 67, no. 4, pp. 789–792, 2006.
[180]  G. Lin, G. Wang, L. Banie et al., “Treatment of stress urinary incontinence with adipose tissue-derived stem cells,” Cytotherapy, vol. 12, no. 1, pp. 88–95, 2010.
[181]  Y. Huang, A. W. Shindel, H. Ning et al., “Adipose derived stem cells ameliorate hyperlipidemia associated detrusor overactivity in a rat model,” Journal of Urology, vol. 183, no. 3, pp. 1232–1240, 2010.
[182]  H. Ning, G. Liu, G. Lin, R. Yang, T. F. Lue, and C. Lin, “Fibroblast growth factor 2 promotes endothelial differentiation of adipose tissue-derived stem cell,” The Journal of Sexual Medicine, vol. 6, no. 4, pp. 967–979, 2009.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413