Abstract
Evidence has emerged that stem cells represent a promising therapeutic tool for tissue engineering and regenerative medicine. Thus, identifying functional markers for selecting stem cells capable of superior self-renewal and pluripotency (or multipotency) and maintaining stem cell identity under appropriate culture conditions are critical for guiding the use of stem cells toward clinical applications. Many investigations have implicated the insulin-like growth factor 1 receptor (IGF1R) signaling in maintenance of stem cell characteristics and enhancement of stem cell therapy efficacy. IGF1R-expressing stem cells display robust pluripotent or multipotent properties. In this review, we summarize the essential roles of IGF1R signaling in self-renewal, pluripotency (or multipotency), and therapeutic efficacy of stem cells, including human embryonic stem cells, neural stem cells, cardiac stem cells, bone marrow mesenchymal stem cells, placental mesenchymal stem cells, and dental pulp mesenchymal stem cells. Modifying IGF1R signaling may thus provide potential strategies for maintaining stem cell properties and improving stem-cell-based therapeutic applications.
Keywords: insulin-like growth factor 1 receptor, multipotency, pluripotency, self-renewal, stem cells
Introduction
Stem cells are biological cells that have the capacity to undergo unlimited numbers of either symmetrical or asymmetrical cell divisions to maintain the stem cell population (self-renewal) as well as produce a broad array of differentiated cell types found in the organism1. There are two major types of stem cells: pluripotent embryonic stem cells (ESCs), which are derived from blastocyst-stage embryos and can generate all types of differentiated cells found in the embryonic tissues2; and multipotent adult stem cells, which are obtained inside different types of tissues and are capable of producing some types of differentiated cells in the organism3, such as the mesenchymal stem cells (MSCs) that are isolated from the connective tissue surrounding other tissues and organs4. Their unique self-renewal ability and multi-lineage potential make stem cells a promising tool for a wide variety of medical therapies (regenerative medicine) such as bone marrow transplantation, myocardial repair, bone regeneration, and nerve regeneration5–7. To guide the use of stem cells toward clinical applications, a key issue to be addressed is the identification and maintenance of stem cells capable of both robust self-renewal and pluripotency (or multipotency) in vitro before in vivo transplantation.
Insulin-like growth factor 1 receptor (IGF1R) is a cell-surface receptor tyrosine kinase that can bind its cognate ligands IGF1 and IGF2 to activate two principle downstream signaling pathways – the phosphoinositide 3-kinase (PI3 K)/AKT and the RAS/mitogen activated protein kinase (MAPK) pathways – to promote cell proliferation, differentiation, migration, and survival, and inhibit apoptosis8–10. Several studies suggest that IGF1R is highly expressed when stem cells are exposed to conditions favorable for self-renewal and pluripotency (or multipotency); the expression of IGF1R recognizes stem cells with superior self-renewal, pluripotency (or multipotency), and therapeutic potential. Activation of IGF1R signaling by autocrine, paracrine, or inter-receptor cross-talk regulations contributes to maintaining the self-renewal and pluripotent (or multipotent) capacities of stem cells. In this review, we provide a quick insight into the essential roles of IGF1R signaling in maintaining stem cell characteristics, and highlight modification of IGF1R signaling as an applicable strategy for improving stem cell-based therapy for human diseases, including heart failure, neurodegenerative diseases, and bone disorders.
Igf1R Signaling Establishes the Stem Cell Niche for Self-Renewal and Pluripotency of Human Embryonic Stem Cells (hESCs)
Human embryonic stem cells are the cells isolated from the inner cell mass of human embryos that are in the blastocyst stage of development11. Although several factors have been identified to play a role in supporting the culture and maintenance of hESCs, including basic fibroblast growth factor (bFGF)12, transforming growth factor β113, activin A14, neurotrophins15, Wnt/β-catenin signaling16, platelet-derived growth factor, and sphingosine-1-phosphate17, little is known about the cell-surface receptors that are activated under conditions supportive of hESC self-renewal.
A report by Wang et al. revealed that when cultured in mouse embryonic fibroblast-conditioned media that support the propagation of undifferentiated hESCs, hESCs displayed prominent tyrosine phosphorylation of IGF1R18. Selective disruption of IGF1R signaling by IGF1R-blocking monoclonal antibody or IGF1R-targeted shRNA severely inhibited hESC proliferation and promoted apoptosis, indicating that IGF1R signaling is required for the self-renewal of pluripotent hESCs18. Furthermore, Bendall et al. clarified that the activation of IGF1R signaling in hESCs depends on a dynamic interplay between hESCs and hESCs-derived fibroblast-like cells19. The hESC-derived fibroblast-like cells were produced by hESCs themselves and acted as a supportive niche via production of IGF2 through a bFGF-dependent autocrine regulation loop19. As a ligand binding to IGF1R, IGF2 has a direct role in sustaining self-renewal and pluripotent properties of hESCs via activation of IGF1R signaling19. Taken together, these reports demonstrate that IGF1R signaling is essential for the acquisition and maintenance of stemness properties of hESCs.
In addition, IGF1R signaling has been implicated in regulating pluripotent ability of hESCs. Magner et al. reported that the expression of both IGF1 and IGF2 and the phosphorylation of IGF1R increased during hepatocyte differentiation from hESCs20. Selective inhibition of IGF1R signaling by small-molecule IGF1R kinase inhibitor or IGF1R-targeted shRNA substantially impaired hepatocyte differentiation, supporting that IGF1R signaling plays an important role in hepatocyte differentiation from hESCs20. Activation of the PI3K/AKT pathway, but not the RAS/MAPK pathway, by IGF1R signaling enhanced the expression of hepatocyte nuclear factor 1 (HNF1) and HNF4 to regulate hepatocyte differentiation from hESCs20. Furthermore, McDevitt et al. reported that IGF1R signaling induced proliferation of cardiomyocytes derived from hESCs21. Blocking of IGF1R by monoclonal antibody attenuated cardiomyocyte proliferation, while addition of IGF1 or IGF2 recombinant protein promoted cardiomyocyte proliferation in a dose-dependent manner21. The proliferation of cardiomyocytes was mediated primarily through the PI3K/AKT pathway downstream of IGF1R signaling21.
Igf1R Signaling Contributes to Human Neural Stem Cell (hNSC)-Mediated Neuroprotection for Amyotrophic Lateral Sclerosis (ALS)
ALS is a lethal neurodegenerative disease that results in loss of motor neurons, leading to rapidly progressive muscular paralysis22. To date, there are no effective treatments for ALS. hNSCs are adult stem cells that are isolated from the human brain and are capable of neural differentiation23. Several clinical trials have supported the use of hNSCs as a promising approach for treating ALS24–27. Enhancing hNSC function may thus increase the benefit of hNSCs-mediated ALS therapy.
Mechanistic investigations of hNSC-mediated neuroprotection revealed that hNSCs produced several neuroprotective growth factors, including vascular endothelial growth factor, brain-derived neurotrophic factor, and IGF1, following intraspinal transplantation in rat and mouse models of ALS, contributing to motor neuron generation, delayed clinical onset, and prolonged life spans28,29. Of these growth factors, IGF1 is the most abundantly expressed.
Reports by Lunn et al. demonstrated that exogenous treatment of IGF1 in hNSC cultures enhanced hNSC neural differentiation and promoted neurite outgrowth in both neurite number and length; the IGF1-stimulated hNSC neurite outgrowth could be abolished by IGF1R inhibitor treatment30. Higher levels of autocrine IGF1 expression in hNSCs consistently increased potential of hNSC migration, stimulated production of glial-derived neurotrophic factors, and induced neural differentiation from hNSCs31. Furthermore, either exogenous treatment or autocrine production of IGF1 augmented the neuroprotective potential of hNSCs and increased motor neuron survival after glutamate exposure in a model of excitotoxic cell death; the IGF1-conferred neuroprotective effect of hNSCs could be abrogated by IGF1R inhibitor treatment30,31. Collectively, these reports support that IGF1R signaling plays an important role in hNSC-mediated neuroprotection and may contribute to the therapeutic benefit of hNSCs for ALS.
Igf1R Signaling Recognized Human Cardiac Stem Cells (hCSCs) with Superior Therapeutic Efficacy for Myocardial Regeneration
Human cardiac stem cells are adult stem cells that are obtained from the human heart and have a tendency to differentiate into cardiac myocytes and vessels32. Considering that age and coronary artery disease may have adverse effects on the function of hCSCs33,34, it is important for tissue regeneration therapy to identify hCSCs with high self-renewal capacity and ability to form myocytes and vessels within the failing heart.
A report by D’Amario et al. showed that the expression of IGF1R in hCSCs identified a pool of hCSCs that exhibited longer telomere length, stronger telomerase activity, enhanced cell proliferation, and decreased apoptosis, whereas absence of IGF1R led to increased apoptosis35. IGF1R-expressing hCSCs produced both IGF1 and IGF2, which supported stem cell proliferation and promoted myocyte differentiation35. Furthermore, IGF1R-positive hCSCs improved cardiomyogenesis and vasculogenesis in a rat model of myocardial infarction; stimulation of IGF1R-positive hCSCs with IGF2 resulted in the development of more mature cardiomyocytes and superior regeneration of ventricular structure35. These results indicate that an IGF1R-positive hCSC subset is an ideal candidate cell for the treatment of human heart failure.
In addition, IGF1 has been known to be a key cardioprotective cytokine that through binding to IGF1R activates IGF1R downstream prosurvival pathways and improves postischemic cardiac function36. Jackson et al. reported that genetically enhancing the paracrine production of IGF1 by transplanted hCSCs promoted hCSCs and cardiomyocyte survival and improved hCSC-mediated myocardial repair in an immunodeficient mouse model of myocardial ischemia, supporting an important role of IGF1R signaling in hCSCs function37.
Igf1R Signaling Promotes Human Bone Marrow Mesenchymal Stem Cell (hBMMSC)-Mediated Myocardial Repair and Bone Formation
Human bone marrow mesenchymal stem cells are MSCs isolated from human bone marrow and are capable of differentiating into several cell types, including cardiomyocytes and vascular endothelial cells38,39. In addition to the application of hCSCs in myocardial regeneration, mentioned above, transplantation of hBMMSCs is also shown to be an attractive approach for myocardial repair. The transplanted hBMMSCs can improve angiogenesis and cardiac function in rat models of heart failure through their ability not only to differentiate into cardiomyocytes and vascular endothelial cells, but also to supply large amounts of angiogenic, anti-apoptotic, and mitogenic factors40–42.
IGF1 has been shown to enhance the migratory response of MSCs to the stromal cell-derived factor-1α (SDF-1α), a potent chemoattractant of stem cells, through activation of the IGF1R downstream PI3K/AKT signaling to increase the expression levels of the SDF-1α receptor, C-X-C motif chemokine receptor 4 (CXCR4)43. Additionally, SDF-1α plays a significant role in modulating stem cell functions via activating molecular pathways of cell growth, proliferation, and survival44. A report by Haider et al. revealed that hBMMSCs, which were transgenically overexpressed with IGF1, showed increased CXCR4 expression with a concomitant increase in SDF-1α production45. After transplantation in a rat model of permanent coronary artery occlusion, the IGF1-overexpressing hBMMSCs accelerated hBMMSC mobilization and retention into the infarcted heart via paracrine activation of SDF-1α/CXCR4 signaling to promote myocardial repair45.
In addition, hBMMSCs can differentiate into chondrocytes and osteoblasts. Longobardi et al. reported that IGF1 is a key factor to promote differentiation of hBMMSCs into chondrocytes by stimulating proliferation, regulating apoptosis, and inducing expression of chondrocyte markers46. The effect of IGF1 on hBMMSC chondrogenesis was mediated by IGF1R downstream MAPK signaling46. Furthermore, IGF1 is the most abundant growth factor in the bone matrix47. A report by Xian et al. showed that IGF1 plays a crucial role in maintaining bone mass through stimulating osteoblastic differentiation of hBMMSCs during bone remodeling, which is mediated by activation of the PI3K/AKT signaling downstream of IGF1R48.
Igf1R Signaling Maintains Self-Renewal and Multipotent Properties of Human Placental Mesenchymal Stem Cells (hPMSCs)
Different parts of the human placenta (including chorionic villi, membranes, umbilical cord, chorioallantois, and amniotic fluid) have been shown as a readily available source of MSCs, termed hPMSCs49. hPMSCs are multipotent and can differentiate into a variety of cell types, including cartilage, bone, endothelial, adipose, muscle, or neuronal lineages50–52. Because studies have shown that hPMSCs have significantly less or no allo- or xenogeneic immune responses53,54, hPMSCs offer great promise for regenerative therapy and tissue engineering. For this purpose, hPMSCs need to be maintained in culture conditions that support their self-renewal and multipotent properties.
Studies have suggested both IGF1 concentration and low-oxygen tension as important regulators for hPMSC physiology in vivo 55,56. Reports by Youssef et al. revealed that exogenous treatment of IGF1 in hPMSC cultures promoted hPMSC proliferation in a dose-dependent manner; the IGF1-mediated hPMSC proliferation was further increased by low-oxygen tension57,58. Furthermore, hPMSC multipotency was also maintained by IGF1 and low-oxygen tension, as shown by increased expression of the ESC marker OCT4 in hPMSCs57,58. Both the proliferation and multipotency of hPMSCs mediated by IGF1 and low-oxygen tension were dependent on IGF1R signaling because inhibition of IGF1R signaling by IGF1R neutralizing antibody or IGF1R-targeted siRNA diminished the proliferation and multipotency of hPMSC in the presence of IGF1 and low-oxygen tension57,58. Overall, these reports indicate that culturing hPMSCs in conditions with IGF1 under low-oxygen tension is critical to maintaining hPMSC multipotency prior to preparation for regenerative therapies.
Igf1R Signaling Enhances Human Dental Pulp Mesenchymal Stem Cell (hDPSC)-Mediated Neuroprotection for Cerebral Hypoxia-Ischemia
Compared with isolation from other sources of MSCs, human dental pulp is regarded as a readily accessible source for MSCs, termed hDPSCs59. hDPSCs could be noninvasively isolated from teeth routinely extracted in the clinic and discarded as medical waste. Moreover, hPDSCs show multipotent capability to differentiate into osteoblasts, odontoblasts, adipocytes, and neural cells60, supporting hPDSCs as a useful source for stem-cell-based therapies.
For the therapeutic applications of MSCs, it is important to identify multipotent markers for selecting MSCs that retain potent self-renewal and multipotent abilities and to maintain the selected MSCs under appropriate culture conditions before in vivo transplantation. Reports by Lee et al. and Chiu et al. revealed that an IGF1R-expressing subpopulation in hDPSCs exhibited both self-renewal and multipotent properties61,62. Importantly, IGF1R expression could be optimally maintained in hPDSCs when they were cultured in 2% human umbilical cord blood serum (hUCBS) in contrast to that in 10% fetal calf serum (FCS)61. Human umbilical cord blood serum contained higher amount of IGF1 compared to FCS, hence triggering a sustained activation of IGF1R signaling61. Also, hDPSC-secreted IGF1 interacted with IGF1R through an autocrine signaling pathway to maintain hDPSC self-renewal62. Furthermore, IGF1 increased expression of CXCR4, a receptor for SDF-1α. Bidirectional cross-talk between IGF1R/IGF1 and CXCR4/SDF-1α signaling synergistically strengthened the activation of IGF1R signaling, contributing to the maintenance of hDPSC stemness61.
In rats with neonatal hypoxia-ischemia, IGF1R-positive hDPSC transplantation to the brain promoted neurite regeneration and improved neurological function through enhancing glucose metabolic activity, inducing angiogenesis and anti-inflammatory effects, increasing anti-apoptotic protein expression, and facilitating cerebral blood flow61,62. In summary, these reports suggest that transplantation of IGF1R-positive hDPSCs is a feasible therapeutic strategy for neurodegenerative diseases.
Conclusion
Although clinical application of stem cells raises some ethical and safety concerns63, results of completed and ongoing clinical trials suggest that stem cells hold great promise in the treatment of a number of human diseases, including degenerative, autoimmune, and genetic disorders64,65. For the purpose of achieving better applications of stem cells in tissue regeneration therapy, it is necessary to isolate highly enriched pluripotent stem cells and maintain their stemness properties in vitro before in vivo transplantation. This review emphasizes that IGF1R signaling is an ideal functional marker for identifying stem cells with superior self-renewal and pluripotent capacities. Moreover, modulating IGF1R signaling activity is a promising strategy to maintain stem cell identity and improve stem cell therapy efficacy. Considering the concern that IGF1R signaling is also implicated in cancer stemness and chemoresistance66,67, the stem cells whose IGF1R signaling activity was modified should be appropriately and adequately differentiated into the tissue-specific cell types in vitro before in vivo transplantation, minimizing the potential side effects of tissue regeneration therapy.
Footnotes
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by grants from the Ministry of Science and Technology, Taiwan (grant number MOST 105-2320-B-039-066).
References
- 1. Sobhani A, Khanlarkhani N, Baazm M, Mohammadzadeh F, Najafi A, Mehdinejadiani S, Sargolzaei Aval F. Multipotent stem cell and current application. Acta Med Iran. 2017;55(1):6–23. [PubMed] [Google Scholar]
- 2. Ohtsuka S, Dalton S. Molecular and biological properties of pluripotent embryonic stem cells. Gene Ther. 2008;15(2):74–81. [DOI] [PubMed] [Google Scholar]
- 3. Tweedell KS. The adaptability of somatic stem cells: a review. J Stem Cells Regen Med. 2017;13(1):3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant. 2011;20(1):5–14. [DOI] [PubMed] [Google Scholar]
- 5. Mahla RS. Stem cells applications in regenerative medicine and disease therapeutics. Int J Cell Biol. 2016;2016:6940283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Levy M, Boulis N, Rao M, Svendsen CN. Regenerative cellular therapies for neurologic diseases. Brain Res. 2016;1638(Pt A):88–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Duelen R, Sampaolesi M. Stem cell technology in cardiac regeneration: a pluripotent stem cell promise. EBioMedicine. 2017;16:30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Girnita L, Worrall C, Takahashi S, Seregard S, Girnita A. Something old, something new and something borrowed: Emerging paradigm of insulin-like growth factor type 1 receptor (IGF-1R) signaling regulation. Cell Mol Life Sci. 2014;71(13):2403–2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of insulin-like growth factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience. 2016;325:89–99. [DOI] [PubMed] [Google Scholar]
- 10. Liu C, Zhang Z, Tang H, Jiang Z, You L, Liao Y. Crosstalk between IGF-1R and other tumor promoting pathways. Curr Pharm Des. 2014;20(17):2912–2921. [DOI] [PubMed] [Google Scholar]
- 11. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–1147. [DOI] [PubMed] [Google Scholar]
- 12. Xu C, Rosler E, Jiang J, Lebkowski JS, Gold JD, O’Sullivan C, Delavan-Boorsma K, Mok M, Bronstein A, Carpenter MK. Basic fibroblast growth factor supports undifferentiated human embryonic stem cell growth without conditioned medium. Stem Cells. 2005;23(3):315–323. [DOI] [PubMed] [Google Scholar]
- 13. Amit M, Shariki C, Margulets V, Itskovitz-Eldor J. Feeder layer- and serum-free culture of human embryonic stem cells. Biol Reprod. 2004;70(3):837–845. [DOI] [PubMed] [Google Scholar]
- 14. Beattie GM, Lopez AD, Bucay N, Hinton A, Firpo MT, King CC, Hayek A. Activin A maintains pluripotency of human embryonic stem cells in the absence of feeder layers. Stem Cells. 2005;23(4):489–495. [DOI] [PubMed] [Google Scholar]
- 15. Pyle AD, Lock LF, Donovan PJ. Neurotrophins mediate human embryonic stem cell survival. Nat Biotechnol. 2006;24(3):344–350. [DOI] [PubMed] [Google Scholar]
- 16. Dravid G, Ye Z, Hammond H, Chen G, Pyle A, Donovan P, Yu X, Cheng L. Defining the role of Wnt/beta-catenin signaling in the survival, proliferation, and self-renewal of human embryonic stem cells. Stem Cells. 2005;23(10):1489–1501. [DOI] [PubMed] [Google Scholar]
- 17. Pebay A, Wong RC, Pitson SM, Wolvetang EJ, Peh GS, Filipczyk A, Koh KL, Tellis I, Nguyen LT, Pera MF. Essential roles of sphingosine-1-phosphate and platelet-derived growth factor in the maintenance of human embryonic stem cells. Stem Cells. 2005;23(10):1541–1548. [DOI] [PubMed] [Google Scholar]
- 18. Wang L, Schulz TC, Sherrer ES, Dauphin DS, Shin S, Nelson AM, Ware CB, Zhan M, Song CZ, Chen X, Brimble SN, McLean A, Galeano MJ, Uhl EW, D’Amour KA, Chesnut JD, Rao MS, Blau CA, Robins AJ. Self-renewal of human embryonic stem cells requires insulin-like growth factor-1 receptor and ERBB2 receptor signaling. Blood. 2007;110(12):4111–4119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Bendall SC, Stewart MH, Menendez P, George D, Vijayaragavan K, Werbowetski-Ogilvie T, Ramos-Mejia V, Rouleau A, Yang J, Bosse M, Lajoie G, Bhatia M. IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro. Nature. 2007;448(7157):1015–1021. [DOI] [PubMed] [Google Scholar]
- 20. Magner NL, Jung Y, Wu J, Nolta JA, Zern MA, Zhou P. Insulin and IGFs enhance hepatocyte differentiation from human embryonic stem cells via the PI3K/AKT pathway. Stem Cells. 2013;31(10):2095–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. McDevitt TC, Laflamme MA, Murry CE. Proliferation of cardiomyocytes derived from human embryonic stem cells is mediated via the IGF/PI 3-kinase/AKT signaling pathway. J Mol Cell Cardiol. 2005;39(6):865–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, Pagani W, Lodin D, Orozco G, Chinea A. A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int. 2015;6:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yan J, Xu L, Welsh AM, Hatfield G, Hazel T, Johe K, Koliatsos VE. Extensive neuronal differentiation of human neural stem cell grafts in adult rat spinal cord. PLoS Med. 2007;4(2):e39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Feldman EL, Boulis NM, Hur J, Johe K, Rutkove SB, Federici T, Polak M, Bordeau J, Sakowski SA, Glass JD. Intraspinal neural stem cell transplantation in amyotrophic lateral sclerosis: Phase 1 trial outcomes. Ann Neurol. 2014;75(3):363–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Riley J, Glass J, Feldman EL, Polak M, Bordeau J, Federici T, Johe K, Boulis NM. Intraspinal stem cell transplantation in amyotrophic lateral sclerosis: A phase I trial, cervical microinjection, and final surgical safety outcomes. Neurosurgery. 2014;74(1):77–87. [DOI] [PubMed] [Google Scholar]
- 26. Mazzini L, Gelati M, Profico DC, et al. Human neural stem cell transplantation in ALS: Initial results from a phase I trial. J Transl Med. 2015;13:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Glass JD, Hertzberg VS, Boulis NM, Riley J, Federici T, Polak M, Bordeau J, Fournier C, Johe K, Hazel T, Cudkowicz M, Atassi N, Borges LF, Rutkove SB, Duell J, Patil PG, Goutman SA, Feldman EL. Transplantation of spinal cord-derived neural stem cells for ALS: analysis of phase 1 and 2 trials. Neurology. 2016;87(4):392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Xu L, Yan J, Chen D, Welsh AM, Hazel T, Johe K, Hatfield G, Koliatsos VE. Human neural stem cell grafts ameliorate motor neuron disease in SOD-1 transgenic rats. Transplantation. 2006;82(7):865–875. [DOI] [PubMed] [Google Scholar]
- 29. Lee HJ, Kim KS, Ahn J, Bae HM, Lim I, Kim SU. Human motor neurons generated from neural stem cells delay clinical onset and prolong life in ALS mouse model. PLoS One. 2014;9(5):e97518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lunn JS, Pacut C, Backus C, Hong Y, Johe K, Hefferan M, Marsala M, Feldman EL. The pleotrophic effects of insulin-like growth factor-I on human spinal cord neural progenitor cells. Stem Cells Dev. 2010;19(12):1983–1993. [DOI] [PubMed] [Google Scholar]
- 31. Lunn JS, Sakowski SA, McGinley LM, Pacut C, Hazel TG, Johe K, Feldman EL. Autocrine production of IGF-I increases stem cell-mediated neuroprotection. Stem Cells. 2015;33(5):1480–1489. [DOI] [PubMed] [Google Scholar]
- 32. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114(6):763–776. [DOI] [PubMed] [Google Scholar]
- 33. Urbanek K, Torella D, Sheikh F, De Angelis A, Nurzynska D, Silvestri F, Beltrami CA, Bussani R, Beltrami AP, Quaini F, Bolli R, Leri A, Kajstura J, Anversa P. Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure. Proc Natl Acad Sci U S A. 2005;102(24):8692–8697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Chimenti C, Kajstura J, Torella D, Urbanek K, Heleniak H, Colussi C, Di Meglio F, Nadal-Ginard B, Frustaci A, Leri A, Maseri A, Anversa P. Senescence and death of primitive cells and myocytes lead to premature cardiac aging and heart failure. Circ Res. 2003;93(7):604–613. [DOI] [PubMed] [Google Scholar]
- 35. D’Amario D, Cabral-Da-Silva MC, Zheng H, Fiorini C, Goichberg P, Steadman E, Ferreira-Martins J, Sanada F, Piccoli M, Cappetta D, D’Alessandro DA, Michler RE, Hosoda T, Anastasia L, Rota M, Leri A, Anversa P, Kajstura J. Insulin-like growth factor-1 receptor identifies a pool of human cardiac stem cells with superior therapeutic potential for myocardial regeneration. Circ Res. 2011;108(12):1467–1481. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 36. Troncoso R, Ibarra C, Vicencio JM, Jaimovich E, Lavandero S. New insights into IGF-1 signaling in the heart. Trends Endocrinol Metab. 2014;25(3):128–137. [DOI] [PubMed] [Google Scholar]
- 37. Jackson R, Tilokee EL, Latham N, Mount S, Rafatian G, Strydhorst J, Ye B, Boodhwani M, Chan V, Ruel M, Ruddy TD, Suuronen EJ, Stewart DJ, Davis DR. Paracrine engineering of human cardiac stem cells with insulin-like growth factor 1 enhances myocardial repair. J Am Heart Assoc. 2015;4(9):e00210 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM. Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest. 2002;109(3):337–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105(1):93–98. [DOI] [PubMed] [Google Scholar]
- 40. Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, Miller L, Guetta E, Zipori D, Kedes LH, Kloner RA, Leor J. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: Feasibility, cell migration, and body distribution. Circulation. 2003;108(7):863–868. [DOI] [PubMed] [Google Scholar]
- 41. Nagaya N, Kangawa K, Itoh T, Iwase T, Murakami S, Miyahara Y, Fujii T, Uematsu M, Ohgushi H, Yamagishi M, Tokudome T, Mori H, Miyatake K, Kitamura S. Transplantation of mesenchymal stem cells improves cardiac function in a rat model of dilated cardiomyopathy. Circulation. 2005;112(8):1128–1135. [DOI] [PubMed] [Google Scholar]
- 42. Tang YL, Zhao Q, Zhang YC, Cheng L, Liu M, Shi J, Yang YZ, Pan C, Ge J, Phillips MI. Autologous mesenchymal stem cell transplantation induce VEGF and neovascularization in ischemic myocardium. Regul Pept. 2004;117(1):3–10. [DOI] [PubMed] [Google Scholar]
- 43. Li Y, Yu X, Lin S, Li X, Zhang S, Song YH. Insulin-like growth factor 1 enhances the migratory capacity of mesenchymal stem cells. Biochem Biophys Res Commun. 2007;356(3):780–784. [DOI] [PubMed] [Google Scholar]
- 44. Mieno S, Ramlawi B, Boodhwani M, Clements RT, Minamimura K, Maki T, Xu SH, Bianchi C, Li J, Sellke FW. Role of stromal-derived factor-1 alpha in the induction of circulating CD34+CXCR4+ progenitor cells after cardiac surgery. Circulation. 2006;114(suppl 1):I186–I192. [DOI] [PubMed] [Google Scholar]
- 45. Haider H, Jiang S, Idris NM, Ashraf M. IGF-1-overexpressing mesenchymal stem cells accelerate bone marrow stem cell mobilization via paracrine activation of SDF-1alpha/CXCR4 signaling to promote myocardial repair. Circ Res. 2008;103(11):1300–1308. [DOI] [PubMed] [Google Scholar]
- 46. Longobardi L, O’Rear L, Aakula S, Johnstone B, Shimer K, Chytil A, Horton WA, Moses HL, Spagnoli A. Effect of IGF-I in the chondrogenesis of bone marrow mesenchymal stem cells in the presence or absence of TGF-beta signaling. J Bone Miner Res. 2006;21(4):626–636. [DOI] [PubMed] [Google Scholar]
- 47. Seck T, Scheppach B, Scharla S, Diel I, Blum WF, Bismar H, Schmid G, Krempien B, Ziegler R, Pfeilschifter J. Concentration of insulin-like growth factor (IGF)-I and -II in iliac crest bone matrix from pre- and postmenopausal women: relationship to age, menopause, bone turnover, bone volume, and circulating IGFs. J Clin Endocrinol Metab. 1998;83(7):2331–2337. [DOI] [PubMed] [Google Scholar]
- 48. Xian L, Wu X, Pang L, Lou M, Rosen CJ, Qiu T, Crane J, Frassica F, Zhang L, Rodriguez JP, Xiaofeng J, Shoshana Y, Shouhong X, Argiris E, Mei W, Xu C. Matrix igf-1 maintains bone mass by activation of mtor in mesenchymal stem cells. Nat Med. 2012;18(7):1095–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. In’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, Kanhai HH. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22(7):1338–1345. [DOI] [PubMed] [Google Scholar]
- 50. Yen BL, Huang HI, Chien CC, Jui HY, Ko BS, Yao M, Shun CT, Yen ML, Lee MC, Chen YC. Isolation of multipotent cells from human term placenta. Stem Cells. 2005;23(1):3–9. [DOI] [PubMed] [Google Scholar]
- 51. Zhang X, Mitsuru A, Igura K, Takahashi K, Ichinose S, Yamaguchi S, Takahashi TA. Mesenchymal progenitor cells derived from chorionic villi of human placenta for cartilage tissue engineering. Biochem Biophys Res Commun. 2006;340(3):944–952. [DOI] [PubMed] [Google Scholar]
- 52. Abdallah BM, Kassem M. Human mesenchymal stem cells: From basic biology to clinical applications. Gene Ther. 2008;15(2):109–116. [DOI] [PubMed] [Google Scholar]
- 53. Bailo M, Soncini M, Vertua E, Signoroni PB, Sanzone S, Lombardi G, Arienti D, Calamani F, Zatti D, Paul P, Albertini A, Zorzi F, Cavagnini A, Candotti F, Wengler GS, Parolini O. Engraftment potential of human amnion and chorion cells derived from term placenta. Transplantation. 2004;78(10):1439–1448. [DOI] [PubMed] [Google Scholar]
- 54. Chen CP, Liu SH, Huang JP, Aplin JD, Wu YH, Chen PC, Hu CS, Ko CC, Lee MY, Chen CY. Engraftment potential of human placenta-derived mesenchymal stem cells after in utero transplantation in rats. Hum Reprod. 2009;24(1):154–165. [DOI] [PubMed] [Google Scholar]
- 55. Genbacev O, Zhou Y, Ludlow JW, Fisher SJ. Regulation of human placental development by oxygen tension. Science. 1997;277(5332):1669–1672. [DOI] [PubMed] [Google Scholar]
- 56. Ong K, Kratzsch J, Kiess W, Costello M, Scott C, Dunger D. Size at birth and cord blood levels of insulin, insulin-like growth factor I (IGF-I), IGF-II, IGF-binding protein-1 (IGFBP-1), IGFBP-3, and the soluble IGF-II/mannose-6-phosphate receptor in term human infants. The ALSPAC study team. Avon longitudinal study of pregnancy and childhood. J Clin Endocrinol Metab. 2000;85(11):4266–4269. [DOI] [PubMed] [Google Scholar]
- 57. Youssef A, Iosef C, Han VK. Low-oxygen tension and IGF-I promote proliferation and multipotency of placental mesenchymal stem cells (PMSCS) from different gestations via distinct signaling pathways. Endocrinology. 2014;155(4):1386–1397. [DOI] [PubMed] [Google Scholar]
- 58. Youssef A, Han VK. Low oxygen tension modulates the insulin-like growth factor-1 or -2 signaling via both insulin-like growth factor-1 receptor and insulin receptor to maintain stem cell identity in placental mesenchymal stem cells. Endocrinology. 2016;157(3):1163–1174. [DOI] [PubMed] [Google Scholar]
- 59. Nakamura S, Yamada Y, Katagiri W, Sugito T, Ito K, Ueda M. Stem cell proliferation pathways comparison between human exfoliated deciduous teeth and dental pulp stem cells by gene expression profile from promising dental pulp. J Endod. 2009;35(11):1536–1542. [DOI] [PubMed] [Google Scholar]
- 60. d’Aquino R, Graziano A, Sampaolesi M, Laino G, Pirozzi G, De Rosa A, Papaccio G. Human postnatal dental pulp cells co-differentiate into osteoblasts and endotheliocytes: A pivotal synergy leading to adult bone tissue formation. Cell Death Differ. 2007;14(6):1162–1171. [DOI] [PubMed] [Google Scholar]
- 61. Lee HT, Chang HT, Lee S, Lin CH, Fan JR, Lin SZ, Hsu CY, Hsieh CH, Shyu WC. Role of IGF1R(+) MSCS in modulating neuroplasticity via CXCR4 cross-interaction. Sci Rep. 2016;6:32595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Chiu HY, Lin CH, Hsu CY, Yu J, Hsieh CH, Shyu WC. IGF1R+ dental pulp stem cells enhanced neuroplasticity in hypoxia-ischemia model. Mol Neurobiol. 2017;54(10):8225–8241. [DOI] [PubMed] [Google Scholar]
- 63. Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, Armstrong L, Djonov V, Lako M, Stojkovic M. Ethical and safety issues of stem cell-based therapy. Int J Med Sci. 2018;15(1):36–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Volarevic V, Ljujic B, Stojkovic P, Lukic A, Arsenijevic N, Stojkovic M. Human stem cell research and regenerative medicine: present and future. Br Med Bull. 2011;99:155–168. [DOI] [PubMed] [Google Scholar]
- 65. Volarevic V, Erceg S, Bhattacharya SS, Stojkovic P, Horner P, Stojkovic M. Stem cell-based therapy for spinal cord injury. Cell Transplant. 2013;22(8):1309–1323. [DOI] [PubMed] [Google Scholar]
- 66. Farabaugh SM, Boone DN, Lee AV. Role of IGF1R in breast cancer subtypes, stemness, and lineage differentiation. Front Endocrinol (Lausanne). 2015;6:59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Ojo D, Wei F, Liu Y, Wang E, Zhang H, Lin X, Wong N, Bane A, Tang D. Factors promoting tamoxifen resistance in breast cancer via stimulating breast cancer stem cell expansion. Curr Med Chem. 2015;22(19):2360–2374. [DOI] [PubMed] [Google Scholar]