Cell Transplantation, Vol. 24, pp. 487–492, 2015 Printed in the USA. All rights reserved. Copyright © 2015 Cognizant Comm. Corp.

0963-6897/15 $90.00 + .00 DOI: http://dx.doi.org/10.3727/096368915X686940 E-ISSN 1555-3892 www.cognizantcommunication.com

Review Adipose Tissue-Derived Stem Cells in Neural Regenerative Medicine Da-Chuan Yeh,*1 Tzu-Min Chan,†‡1 Horng-Jyh Harn,§¶ Tzyy-Wen Chiou,# Hsin-Shui Chen,**†† Zung-Sheng Lin,‡‡ and Shinn-Zong Lin§§¶¶##*** *Department of Internal Medicine, China Medical University Beigan Hospital, Yunlin, Taiwan †Department of Medical Education and Research, China Medical University Beigan Hospital, Yunlin, Taiwan ‡Department of Medical Education and Research, China Medical University-An-Nan Hospital, Tainan, Taiwan §Department of Medicine, China Medical University, Taichung, Taiwan ¶Department of Pathology, China Medical University Hospital, Taichung, Taiwan #Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan **School of Medicine, China Medical University, Taichung, Taiwan ††Department of Physical Medicine and Rehabilitation, China Medical University Beigang Hospital, Yunlin, Taiwan ‡‡Department of Surgery, China Medical University Beigan Hospital, Yunlin, Taiwan §§Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan ¶¶Graduate Institute of Immunology, China Medical University, Taichung, Taiwan ##Department of Neurosurgery, China Medical University Beigan Hospital, Yunlin, Taiwan ***Department of Neurosurgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan

Adipose tissue-derived stem cells (ADSCs) have two essential characteristics with regard to regenerative medicine: the convenient and efficient generation of large numbers of multipotent cells and in vitro proliferation without a loss of stemness. The implementation of clinical trials has prompted widespread concern regarding safety issues and has shifted research toward the therapeutic efficacy of stem cells in dealing with neural degeneration in cases such as stroke, amyotrophic lateral sclerosis, Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, cavernous nerve injury, and traumatic brain injury. Most existing studies have reported that cell therapies may be able to replenish lost cells and promote neuronal regeneration, protect neuronal survival, and play a role in overcoming permanent paralysis and loss of sensation and the recovery of neurological function. The mechanisms involved in determining therapeutic capacity remain largely unknown; however, this concept can still be classified in a methodical manner by citing current evidence. Possible mechanisms include the following: 1) the promotion of angiogenesis, 2) the induction of neuronal differentiation and neurogenesis, 3) reductions in reactive gliosis, 4) the inhibition of apoptosis, 5) the expression of neurotrophic factors, 6) immunomodulatory function, and 7) facilitating neuronal integration. In this study, several human clinical trials using ADSCs for neuronal disorders were investigated. It is suggested that ADSCs are one of the choices among various stem cells for translating into clinical application in the near future. Key words: Adipose tissue-derived stem cells (ADSCs); Stroke; Amyotrophic lateral sclerosis; Parkinson’s disease; Alzheimer’s disease; Huntington’s disease; Cavernous nerve injury

ADIPOSE TISSUE-DERIVED STEM CELLS (ADSCs)

have previously been associated with differentiation include cluster of differentiation 29 (CD29), CD44, CD73, CD90, and CD105 as well as human leukocyte antigen class I. Other distinguishing surface marker expression profiles of ADSCs are likely to include CD9+, CD10+, CD13+, CD49d+, CD49e+, CD54+, CD55+, CD71+, CD106+,

ADSCs are mesenchymal stem cells (MSCs) that can be isolated from the abdominal subcutaneous, intra-articular and visceral stations, and ectopic fat tissue using minimally invasive methods (4,14). Notable markers of ADSCs that

Received December 9, 2014; final acceptance January 27, 2015. Online prepub date: February 2, 2015. 1 These authors provided equal contribution to this work. Address correspondence to Prof. Shinn-Zong Lin, M.D., Ph.D., Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan, ROC. Tel: +886-4-22052121, ext. 6034; Fax: +886-4-220806666; E-mail: [email protected]

487

488

CD146+, CD166+, and STRO-1+ (5,61). The differentiation of ADSCs suggests that they could be induced into a wide variety of tissue-specific lineages, including adipocytes, osteoblasts, chondrocytes, hepatocytes, myocytes, and epithelial and neuronal cells (4). A growing body of evidence has indicated the therapeutic capacity of ADSCs in dealing with a variety of issues associated with stroke, amyotrophic lateral sclerosis (ALS), Parkinson’s disease (PD), autoimmune diseases, multiple sclerosis, polymyositis, dermatomyositis, and rheumatoid arthritis (15). In addition, ADSCs have undergone intensive scrutiny with regard to their safety, including karyotype analysis, telomerase activity, and cell proliferation (38,44). However, Yu et al. (58) indicated that MSCs could favor tumor growth in vivo; thus, it is necessary to test the long-term safety of ADSCs. Ra et al. (39) tested the toxicity of intravenous infusion of ADSCs in animals and humans, and the results showed no side effects and no evidence of tumor development. Thus, the safety, therapeutic potential, and clinical applicability of ADSCs place them among the most promising resources in the field of regenerative therapy. CHRONIC STROKE The syndromes of stroke include the loss of nerve function with sudden hemiplegia, aphasia, and numbness in hands and feet, most of which are caused by the blockage of blood vessels in parts of the brain (46,47). Stroke can be classified as either ischemic involving the loss of normal blood supply or hemorrhagic (commonly known as hyperemia), which is determined by the type of blood vessel injury in brain (22,41). Acute stroke is defined as neurological deficits of vascular origin occurring within a period of a few hours. In contrast, chronic stroke is normally associated with attacks lasting for a prolonged duration (51). Most current regimens used in the treatment of acute stroke provide only a partial restoration of the injured region. The results showed that the administration of ADSCs intracerebroventricularly (ICV) and intravenously (IV) into the injured region have potential therapeutic effects and tissue regeneration ability in rodent models (4,18,19). The application of ADSCs to the problem of chronic stroke involves finding a workable strategy with which to promote functional neuron regeneration. In the chronic stroke region, the scattered blood vessels usually are not able to support nutrients for maintaining cell survival; thus, vascular endothelial growth factor (VEGF) might be a possible mechanism that protects neurons from cell death (11). The administration of ADSCs could induce neuronal differentiation and stimulate brain repair markers associated with neurogenesis to promote functional recovery from chronic stroke (6,18,43). Nonetheless, this could cause glial scarring at the infarct boundary in cases involving the excess proliferation or hypertrophy of glial cells, such as astrocytes, microglia,

YEH ET AL.

and oligodendrocytes (3,9). The application of ADSCs for the treatment of stroke is meant to reduce the generation of glial fibrillary acidic protein-positive cells, thereby preventing nerve fibrosis in the brain (56). In addition, neuronal recovery is usually accompanied by a reduction in brain injury-derived apoptosis as well as natural repair responses that are activated after brain injury (18). ADSCs also enhance the survival and/or differentiation of neuronal cells via neurotrophic factors (25,36). Another possible mechanism underlying the protective role of ADSCs may be through an immunomodulatory mechanism that displayed increased interleukin-10 and reduced tumor necrosis factor-a in middle cerebral artery occlusion rats (10). In summary, these results indicate that the administration of ADSCs in rodent stroke models can have significantly positive effects on neuronal function. ALS ALS is a neuromuscular degenerative disease involving the death of lower and upper motor neurons (12). The primary clinical manifestation is a gradual weakening and atrophy of muscle tissue, resulting in progressive failure of the neuromuscular system and eventual death (7). The treatment modality remains an unsolved mystery, and no effective therapy is currently available for this disease (24). ADSC transplantation has been used in various diseases associated with neurological deficits; therefore, it may be a therapeutic agent for ALS. It has been reported that ADSCs can differentiate into neuron-like cells, and delivery of ADSCs through ICV or IV prior to the appearance of clinical symptoms has been shown to delay the onset of the disease and extend life spans in a superoxide dismutase 1 G93A-mutated (SOD1G93A) ALS mouse model (25). Following transplantation, increased levels of neurotrophic factors, such as nerve growth factor, brain-derived neurotrophic factor, insulin-like growth factor, and VEGF, as well as decreased apoptotic cell death were detected in the spinal cord of the ALS mice (25). Another report showed similar outcomes with significant upregulation of glial cell-derived neurotrophic factor and basic fibroblast growth factor, indicating that ADSCs may play a role in neuroprotection as well as the mitigation of symptoms (32). These results from animal models provide valuable data to support future trials in ALS patients. PARKINSON’S DISEASE The motor symptoms of PD are due to programmed cell death of dopaminergic neurons in the substantia nigra, as well as interruptions in nerve conduction paths (16). The main cause of PD is mutations in the a-synuclein gene resulting in protein aggregation and the formation of Lewy bodies, Lewy neuritis, and cytotoxicity, which may induce spontaneous cell death (23). Previous studies have reported on the use of in vitro cell culture systems to test the ability

APPLICATION OF ADIPOSE TISSUE-DERIVED STEM CELLS

489

of proteins secreted from ADSCs to recover damage caused by 6-hydroxydopamine (6-OHDA) (17). The results demonstrate that ADSC-conditioned media is able to recover 6-OHDA-induced reactive oxygen species and neurotoxicity in rat mesencephalic neurons and cerebellar granule neurons, and directly attenuate H2O2-induced neuronal death (17). In addition, ADSC-conditioned media has been shown to promote neurite regeneration in PC12 cells by increasing bone morphogenetic protein 2 and FGF2 (34). Moreover, ADSC implantation into hemiparkinsonian rhesus monkeys, generated by methyl-4-phenyl-1, 2, 3, 6tetrahydropyridine (MPTP), has been shown to have in vivo neuroprotective effects (60). Improved treatment protocols, such as an in vitro strategy for neuronal differentiation treated with LIM homeobox transcription factor 1a (LMX1A) and neurturin, have shown improvements in behavior and symptom amelioration in monkey models (60). In the future, ADSCs may provide a valuable resource in the transplantation of autologous cells for the treatment of PD.

by the misfolded Huntingtin mutant protein (35). The transplantation of ADSCs into R6/2 transgenic mice with Huntington’s disease was shown to promote the secretion of multiple paracrine growth factors (21). The effect of the protective factors is to prevent apoptotic phenomena and recover behavioral deficiencies in animal models. An alternative method has been proposed from the same research group in which cell extracts were used, instead of the direct transplantation of ADSCs, for intraperitoneal injection (20). Those results demonstrated significant improvements in behavior and the rotarod test as well as reductions in striatal atrophy and abnormal Huntington aggregation, which may be regulated via the cyclic adenosine monophosphate-responsive element-binding proteinperoxisome proliferator-activated receptor g, coactivator 1 a pathway. Other animal models have been used to develop strategies to deal with Huntington diseases (33). According to the results of rodent models, ADSCs may provide a curative autologous cell-based therapy for Huntington’s disease.

ALZHEIMER’S DISEASE

CAVERNOUS NERVE INJURY

Although Alzheimer’s disease is the most common type of dementia, little is known with regard to the symptoms of late onset Alzheimer’s (50). Several opposing hypotheses have been forwarded to explain the cause of the dementia, including cholinergic (13), amyloid (28), tau (8), age-related myelin breakdown (2), and oxidative stress (48). Recent studies have reported that ADSCs are a promising new cell source for regenerative cures, capable of ameliorating the neuropathological deficits associated with Alzheimer’s disease (31). Ma and his colleagues demonstrated the efficacy of intracerebral administration of ADSCs in the recovery of spatial learning/memory ability in amyloid precursor protein/presenilin 1 (APP/PS1) double transgenic mice (31). In addition, the IV administration of ADSCs has also been shown to have palliative effects on the symptoms of dementia and therapeutic effects in Alzheimer’s disease mouse models (26). Perez-Gonzalez et al. investigated the issue of neurogenesis in the adult hippocampus (37), the results of which indicated an increase in the proliferation of neuronal precursors and survival of neurons in APP/PS1 double transgenic mice. In summary, the transplantation of ADSCs potentially has beneficial effects in the prevention of pathological deterioration associated with Alzheimer’s disease.

The cavernous nerve is in line with the posterolateral aspects of the prostate, which in turn provides the majority of autonomic input to erectile tissue. Injuries resulting from prostatectomy complications typically result in erectile dysfunction (52,57). Recently, much research has sought to develop 1) neurotrophic and neuroprotective agents to restore erectile function and 2) intracavernosal or IV ADSC injections to regenerate cavernous nerves (1,30,54). Previous studies demonstrated that ADSCs secrete neurotrophic factors (which promote cavernous nerve regeneration) and differentiate into Schwann cells (which play pivotal roles in peripheral nerves). Furthermore, other researchers have shown that ADSCs have therapeutic potential to treat postprostatectomy erectile dysfunction (1,27,59). For example, You et al. found that intracavernosal injections of human ADSCs effectively restored penile erectile function in a rat model of cavernous nerve injury (57), and Xu et al. found that using ADSC-based microtissues to treat postprostatectomy erectile dysfunction yielded significant benefits (54). Nonetheless, human clinical trials are necessary to further investigate therapeutic effects and safety issues associated with ADSC treatments.

HUNTINGTON’S DISEASE Huntington’s is a rare, inherited, late onset neurodegenerative genetic disorder, characterized by defects in muscular coordination and cognitive decline (42). The pathology mechanisms include activated apoptotic signaling, abnormal glutamine function, the impairment of energy production, and altered gene expression, due to cellular toxicity caused

TRAUMATIC BRAIN INJURY Traumatic brain injury (TBI), also known as intracranial injury, occurs when external trauma causes the loss of motor and cognitive functions in the brain (49). In recent studies, stem cell treatments have been shown to effectively restore physiological functions in animal models of TBI (45,53). Furthermore, findings suggest that ADSCs may present an attractive cell-based therapy for regenerative medicine and wound healing in TBI (55). Specifically,

490

YEH ET AL.

the authors showed that adult ADSCs can be induced to differentiate into functional endothelial progenitor cells, which have beneficial effects on cell therapy. Moreover, Tajiri et al. treated F344 rats with an IV injection of hADSCs, conditioned media, or a vehicle (unconditioned media) 3 h following TBI (49). In that study, rats showed significant amelioration of motor and cognitive functions as well as significant reduction in cortical damage and hippocampal cell loss. Findings from that study and others have advanced scientific understanding related to potential therapeutic effects of ADSCs in TBI and have provided pivotal guidance for future human clinical trials. FUTURE RESEARCH EXPECTATIONS ADSCs are applicable to the treatment of human neurodegenerative disorders, due in part to their availability and safety (15). Future development will require improvements in cell manipulation technology. Besides, the development of stem cell treatments will require a safe method of visual tracking in order to avoid the need for toxic imaging and invasive diagnostic procedures (29,40). A study with the tracking and visualization of cells or the pharmacokinetics of small molecule drugs, verifying the safety and therapeutic benefits of proposed experiments is crucial to gaining approval for human clinical trials. In addition, developing a sterile system that requires minimal upkeep for the generation of nucleated cells could have considerable clinical applicability. Furthermore, auxiliary methods could enhance neurogenesis from endogenous progenitors, while drugs or chemical compounds could be included in combinational cures (12,13). In summary, the success of these treatments in animal experiments suggests their efficacy should be further explored in future human clinical trials. ACKNOWLEDGMENTS: This study is supported in part by Taiwan Ministry of Health and Welfare Clinical Trial and Research Center of Excellence (MOHW103-TDU-B-212-113002), CMUBH R103-012, and CMUBH R103-007. The authors declare no conflicts of interest.

REFERENCES 1. Albersen, M.; Fandel, T. M.; Lin, G.; Wang, G.; Banie, L.; Lin, C. S.; Lue, T. F. 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(10):3331–3340; 2010. 2. Bartzokis, G. Alzheimer’s disease as homeostatic responses to age-related myelin breakdown. Neurobiol. Aging 32(8): 1341–1371; 2011. 3. Beck, H.; Semisch, M.; Culmsee, C.; Plesnila, N.; Hatzopoulos, A. K. Egr-1 regulates expression of the glial scar component phosphacan in astrocytes after experimental stroke. Am. J. Pathol. 173(1):77–92; 2008. 4. Chan, T. M.; Harn, H. J.; Lin, H. P.; Chiu, S. C.; Lin, P. C.; Wang, H. I.; Ho, L. I.; Chuu, C. P.; Chiou, T. W.; Hsieh, A. C.; Chen, Y. W.; Ho, W. Y.; Lin, S. Z. The use of ADSCs as a treatment for chronic stroke. Cell Transplant. 23(4):541–547; 2014.

5. Chan, T. M.; Harn, H. J.; Lin, H. P.; Chou, P. W.; Chen, J. Y.; Ho, T. J.; Chiou, T. W.; Chuang, H. M.; Chiu, S. C.; Chen, Y. C.; Yen, S. Y.; Huang, M. H.; Liang, B. C.; Lin, S. Z. Improved human mesenchymal stem cell isolation. Cell Transplant. 23(4):399–406; 2014. 6. Chen, J.; Tang, Y. X.; Liu, Y. M.; Hu, X. Q.; Liu, N.; Wang, S. X.; Zhang, Y.; Zeng, W. G.; Ni, H. J.; Zhao, B.; Chen, Y. F.; Tang, Z. P. Transplantation of adipose-derived stem cells is associated with neural differentiation and functional improvement in a rat model of intracerebral hemorrhage. CNS Neurosci. Ther. 18(10):847–854; 2012. 7. Ching, J. K.; Elizabeth, S. V.; Ju, J. S.; Lusk, C.; Pittman, S. K.; Weihl, C. C. mTOR dysfunction contributes to vacuolar pathology and weakness in valosin-containing protein associated inclusion body myopathy. Hum. Mol. Genet. 22(6):1167–1179; 2013. 8. Chun, W.; Johnson, G. V. The role of tau phosphorylation and cleavage in neuronal cell death. Front. Biosci. 12:733– 756; 2007. 9. Chung, J. Y.; Kim, W.; Im, W.; Yoo, D. Y.; Choi, J. H.; Hwang, I. K.; Won, M. H.; Chang, I. B.; Cho, B. M.; Hwang, H. S.; Moon, S. M. Neuroprotective effects of adipose-derived stem cells against ischemic neuronal damage in the rabbit spinal cord. J. Neurol. Sci. 317(1–2):40–46; 2012. 10. Du, H. W.; Liu, N.; Wang, J. H.; Zhang, Y. X.; Chen, R. H.; Xiao, Y. C. [The effects of adipose-derived stem cell transplantation on the expression of IL-10 and TNF-alpha after cerebral ischaemia in rats]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 25(11):998–1001; 2009. 11. Fang, Z.; Yang, Q.; Xiong, W.; Li, G.; Xiao, J.; Guo, F.; Li, F.; Chen, A. Neurogenic differentiation of murine adipose derived stem cells transfected with EGFP in vitro. J. Huazhong Univ. Sci. Technolog. Med. Sci. 30(1):75–80; 2010. 12. Ferraiuolo, L.; Kirby, J.; Grierson, A. J.; Sendtner, M.; Shaw, P. J. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat. Rev. Neurol. 7(11):616– 630; 2011. 13. Francis, P. T.; Palmer, A. M.; Snape, M.; Wilcock, G. K. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 66(2):137– 147; 1999. 14. Gimble, J. M.; Bunnell, B. A.; Frazier, T.; Rowan, B.; Shah, F.; Thomas-Porch, C.; Wu, X. Adipose-derived stromal/ stem cells: A primer. Organogenesis 9(1):3–10; 2013. 15. Gir, P.; Oni, G.; Brown, S. A.; Mojallal, A.; Rohrich, R. J. Human adipose stem cells: Current clinical applications. Plast. Reconstr. Surg. 129(6):1277–1290; 2012. 16. Goldstein, D. S. Biomarkers, mechanisms, and potential prevention of catecholamine neuron loss in Parkinson disease. Adv. Pharmacol. 68:235–272; 2013. 17. Gu, H.; Wang, J.; Du, N.; Tan, J.; Johnstone, B.; Du, Y. Adipose stromal cells-conditioned medium blocks 6hydroxydopamine-induced neurotoxicity and reactive oxygen species. Neurosci. Lett. 544:15–19; 2013. 18. Gutierrez-Fernandez, M.; Rodriguez-Frutos, B.; Otero-Ortega, L.; Ramos-Cejudo, J.; Fuentes, B.; Diez-Tejedor, E. Adipose tissue-derived stem cells in stroke treatment: From bench to bedside. Discov. Med. 16(86):37–43; 2013. 19. Ikegame, Y.; Yamashita, K.; Hayashi, S.; Mizuno, H.; Tawada, M.; You, F.; Yamada, K.; Tanaka, Y.; Egashira, Y.; Nakashima, S.; Yoshimura, S.; Iwama, T. Comparison of mesenchymal stem cells from adipose tissue and bone marrow for ischemic stroke therapy. Cytotherapy 13(6):675– 685; 2011.

APPLICATION OF ADIPOSE TISSUE-DERIVED STEM CELLS

20. Im, W.; Ban, J.; Lim, J.; Lee, M.; Lee, S. T.; Chu, K.; Kim, M. Extracts of adipose derived stem cells slows progression in the R6/2 model of Huntington’s disease. PLoS One 8(4): e59438; 2013. 21. Im, W.; Lee, S. T.; Park, J. E.; Oh, H. J.; Shim, J.; Lim, J.; Chu, K.; Kim, M. Transplantation of patient-derived adipose stem cells in YAC128 Huntington’s disease transgenic mice. PLoS Curr. 2:RRN1183; 2010. 22. Kalantri, A.; Kalantri, S. Distinguishing hemorrhagic stroke from ischemic stroke. JAMA 304(12):1327–1328; author reply 1328; 2010. 23. Kalia, L. V.; Kalia, S. K.; McLean, P. J.; Lozano, A. M.; Lang, A. E. alpha-Synuclein oligomers and clinical implications for Parkinson disease. Ann. Neurol. 73(2):155–169; 2013. 24. Kato, A. C.; Vingerhoets, F. J.; Magistris, M. R.; Kuntzer, T.; Burkhard, P. R. [Pathogenic mechanisms of neurodegenerative diseases: Amyotrophic lateral sclerosis]. Rev. Med. Suisse 2(64):1152–1154, 1156–1157; 2006. 25. Kim, K. S.; Lee, H. J.; An, J.; Kim, Y. B.; Ra, J. C.; Lim, I.; Kim, S. U. Transplantation of human adipose tissuederived stem cells delays clinical onset and prolongs life span in ALS mouse model. Cell Transplant. 23(12):1585– 1597; 2014. 26. Kim, S.; Chang, K. A.; Kim, J.; Park, H. G.; Ra, J. C.; Kim, H. S.; Suh, Y. H. The preventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer’s disease mice. PLoS One 7(9):e45757; 2012. 27. Kingham, P. J.; Kalbermatten, D. F.; Mahay, D.; Armstrong, S. J.; Wiberg, M.; Terenghi, G. Adipose-derived stem cells differentiate into a Schwann cell phenotype and promote neurite outgrowth in vitro. Exp. Neurol. 207(2):267–274; 2007. 28. Lauren, J.; Gimbel, D. A.; Nygaard, H. B.; Gilbert, J. W.; Strittmatter, S. M. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 457(7233):1128–1132; 2009. 29. Lee, H. J.; Won, J. H.; Doo, S. H.; Kim, J. H.; Song, K. Y.; Lee, S. J.; Lim, I.; Chang, K. T.; Song, Y. S.; Kim, S. U. Inhibition of collagen deposit in obstructed rat bladder outlet by transplantation of superparamagnetic iron oxidelabeled human mesenchymal stem cells as monitored by molecular magnetic resonance imaging (MRI). Cell Transplant. 21(5):959–970; 2012. 30. Lin, G.; Albersen, M.; Harraz, A. M.; Fandel, T. M.; Garcia, M.; McGrath, M. H.; Konety, B. R.; Lue, T. F.; Lin, C. S. Cavernous nerve repair with allogenic adipose matrix and autologous adipose-derived stem cells. Urology 77(6):1509.e1501–1509.e1508; 2011. 31. Ma, T.; Gong, K.; Ao, Q.; Yan, Y.; Song, B.; Huang, H.; Zhang, X.; Gong, Y. Intracerebral transplantation of adiposederived mesenchymal stem cells alternatively activates microglia and ameliorates neuropathological deficits in Alzheimer’s disease mice. Cell Transplant. 22(Suppl 1):S113–S126; 2013. 32. Marconi, S.; Bonaconsa, M.; Scambi, I.; Squintani, G. M.; Rui, W.; Turano, E.; Ungaro, D.; D’Agostino, S.; Barbieri, F.; Angiari, S.; Farinazzo, A.; Constantin, G.; Del Carro, U.; Bonetti, B.; Mariotti, R. Systemic treatment with adiposederived mesenchymal stem cells ameliorates clinical and pathological features in the amyotrophic lateral sclerosis murine model. Neuroscience 248C:333–343; 2013. 33. Maucksch, C.; Vazey, E. M.; Gordon, R. J.; Connor, B. Stem cell-based therapy for Huntington’s disease. J. Cell. Biochem. 114(4):754–763; 2013.

491

34. Moriyama, M.; Moriyama, H.; Ueda, A.; Nishibata, Y.; Okura, H.; Ichinose, A.; Matsuyama, A.; Hayakawa, T. Human adipose tissue-derived multilineage progenitor cells exposed to oxidative stress induce neurite outgrowth in PC12 cells through p38 MAPK signaling. BMC Cell Biol. 13:21; 2012. 35. Nucifora, F. C. Jr.; Sasaki, M.; Peters, M. F.; Huang, H.; Cooper, J. K.; Yamada, M.; Takahashi, H.; Tsuji, S.; Troncoso, J.; Dawson, V. L.; Dawson, T. M.; Ross, C. A. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science 291(5512): 2423–2428; 2001. 36. Ohta, Y.; Takenaga, M.; Tokura, Y.; Hamaguchi, A.; Matsumoto, T.; Kano, K.; Mugishima, H.; Okano, H.; Igarashi, R. Mature adipocyte-derived cells, dedifferentiated fat cells (DFAT), promoted functional recovery from spinal cord injury-induced motor dysfunction in rats. Cell Transplant. 17(8):877–886; 2008. 37. Perez-Gonzalez, R.; Antequera, D.; Vargas, T.; Spuch, C.; Bolos, M.; Carro, E. Leptin induces proliferation of neuronal progenitors and neuroprotection in a mouse model of Alzheimer’s disease. J. Alzheimers Dis. 24(Suppl 2):17– 25; 2011. 38. Ra, J. C.; Kang, S. K.; Shin, I. S.; Park, H. G.; Joo, S. A.; Kim, J. G.; Kang, B. C.; Lee, Y. S.; Nakama, K.; Piao, M.; Sohl, B.; Kurtz, A. Stem cell treatment for patients with autoimmune disease by systemic infusion of culture-expanded autologous adipose tissue derived mesenchymal stem cells. J. Transl. Med. 9:181; 2011. 39. Ra, J. C.; Shin, I. S.; Kim, S. H.; Kang, S. K.; Kang, B. C.; Lee, H. Y.; Kim, Y. J.; Jo, J. Y.; Yoon, E. J.; Choi, H. J.; Kwon, E. Safety of intravenous infusion of human adipose tissue-derived mesenchymal stem cells in animals and humans. Stem Cells Dev. 20(8):1297–1308; 2011. 40. Raschzok, N.; Langer, C. M.; Schmidt, C.; Lerche, K. H.; Billecke, N.; Nehls, K.; Schluter, N. B.; Leder, A.; Rohn, S.; Mogl, M. T.; Ludemann, L.; Stelter, L.; Teichgraber, U. K.; Neuhaus, P.; Sauer, I. M. Functionalizable silica-based micron-sized iron oxide particles for cellular Magnetic Resonance Imaging. Cell Transplant. 22(11):1959–1970; 2013. 41. Ribo, M.; Rubiera, M.; Pagola, J.; Rodriguez-Luna, D.; Meler, P.; Flores, A.; Alvarez-Sabin, J.; Molina, C. A. Bringing forward reperfusion with oxygenated blood perfusion beyond arterial occlusion during endovascular procedures in patients with acute ischemic stroke. AJNR Am. J. Neuroradiol. 31(10):1899–1902; 2010. 42. Roos, R. A. Huntington’s disease: A clinical review. Orphanet. J. Rare Dis. 5(1):40; 2010. 43. Safford, K. M.; Hicok, K. C.; Safford, S. D.; Halvorsen, Y. D.; Wilkison, W. O.; Gimble, J. M.; Rice, H. E. Neurogenic differentiation of murine and human adipose-derived stromal cells. Biochem. Biophys. Res. Commun. 294(2):371–379; 2002. 44. Safwani, W. K.; Makpol, S.; Sathapan, S.; Chua, K. H. The impact of long-term in vitro expansion on the senescenceassociated markers of human adipose-derived stem cells. Appl. Biochem. Biotechnol. 166(8):2101–2113; 2012. 45. Shear, D. A.; Tate, C. C.; Tate, M. C.; Archer, D. R.; LaPlaca, M. C.; Stein, D. G.; Dunbar, G. L. Stem cell survival and functional outcome after traumatic brain injury is dependent on transplant timing and location. Restor. Neurol. Neurosci. 29(4):215–225; 2011. 46. Shyu, W. C.; Chen, C. P.; Lin, S. Z.; Lee, Y. J.; Li, H. Efficient tracking of non-iron-labeled mesenchymal stem cells with serial MRI in chronic stroke rats. Stroke 38(2):367–374; 2007.

492

47. Shyu, W. C.; Lin, S. Z.; Yang, H. I.; Tzeng, Y. S.; Pang, C. Y.; Yen, P. S.; Li, H. Functional recovery of stroke rats induced by granulocyte colony-stimulating factor-stimulated stem cells. Circulation 110(13):1847–1854; 2004. 48. Su, B.; Wang, X.; Nunomura, A.; Moreira, P. I.; Lee, H. G.; Perry, G.; Smith, M. A.; Zhu, X. Oxidative stress signaling in Alzheimer’s disease. Curr. Alzheimer Res. 5(6): 525–532; 2008. 49. Tajiri, N.; Acosta, S. A.; Shahaduzzaman, M.; Ishikawa, H.; Shinozuka, K.; Pabon, M.; Hernandez-Ontiveros, D.; Kim, D. W.; Metcalf, C.; Staples, M.; Dailey, T.; Vasconcellos, J.; Franyuti, G.; Gould, L.; Patel, N.; Cooper, D.; Kaneko, Y.; Borlongan, C. V.; Bickford, P. C. Intravenous transplants of human adipose-derived stem cell protect the brain from traumatic brain injury-induced neurodegeneration and motor and cognitive impairments: Cell graft biodistribution and soluble factors in young and aged rats. J. Neurosci. 34(1): 313–326; 2014. 50. Tanzi, R. E. A brief history of Alzheimer’s disease gene discovery. J. Alzheimers Dis. 33(Suppl 1):S5–S13; 2013. 51. Thomas, J.; Fitzpatrick, L. Acute ischemic stroke review. J. Neurosci. Nurs. 40(2):69; author reply 69; 2008. 52. Walsh, P. C.; Donker, P. J. Impotence following radical prostatectomy: Insight into etiology and prevention. J. Urol. 128(3):492–497; 1982. 53. Wang, S.; Cheng, H.; Dai, G.; Wang, X.; Hua, R.; Liu, X.; Wang, P.; Chen, G.; Yue, W.; An, Y. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res. 1532:76–84; 2013. 54. Xu, Y.; Guan, R.; Lei, H.; Li, H.; Wang, L.; Gao, Z.; Song, W.; Xin, Z. Therapeutic potential of adipose-derived stem

YEH ET AL.

55.

56.

57.

58. 59.

60.

61.

cells-based micro-tissues in a rat model of postprostatectomy erectile dysfunction. J. Sex. Med. 11(1):2439–2448; 2014. Xue, S.; Zhang, H. T.; Zhang, P.; Luo, J.; Chen, Z. Z.; Jang, X. D.; Xu, R. X. Functional endothelial progenitor cells derived from adipose tissue show beneficial effect on cell therapy of traumatic brain injury. Neurosci. Lett. 473(3): 186–191; 2010. Yang, Y. C.; Liu, B. S.; Shen, C. C.; Lin, C. H.; Chiao, M. T.; Cheng, H. C. Transplantation of adipose tissue-derived stem cells for treatment of focal cerebral ischemia. Curr. Neurovasc. Res. 8(1):1–13; 2011. You, D.; Jang, M. J.; Lee, J.; Suh, N.; Jeong, I. G.; Sohn, D. W.; Kim, S. W.; Ahn, T. Y.; Kim, C. S. 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 73(3):278–286; 2013. Yu, J. M.; Jun, E. S.; Bae, Y. C.; Jung, J. S. Mesenchymal stem cells derived from human adipose tissues favor tumor cell growth in vivo. Stem Cells Dev. 17(3):463–473; 2008. Zhang, H.; Yang, R.; Wang, Z.; Lin, G.; Lue, T. F.; Lin, C. S. Adipose tissue-derived stem cells secrete CXCL5 cytokine with neurotrophic effects on cavernous nerve regeneration. J. Sex. Med. 8(2):437–446; 2011. Zhou, Y.; Sun, M.; Li, H.; Yan, M.; He, Z.; Wang, W.; Lu, S. Recovery of behavioral symptoms in hemi-Parkinsonian rhesus monkeys through combined gene and stem cell therapy. Cytotherapy 15(4):467–480; 2013. Zuk, P. A. The adipose-derived stem cell: Looking back and looking ahead. Mol. Biol. Cell 21(11):1783–1787; 2010.

Adipose tissue-derived stem cells in neural regenerative medicine.

Adipose tissue-derived stem cells (ADSCs) have two essential characteristics with regard to regenerative medicine: the convenient and efficient genera...
184KB Sizes 2 Downloads 13 Views