Intranasal administration of human MSC for ischemic brain injury in the mouse: in vitro and in vivo neuroregenerative functions

Vanessa Donega, Cora H Nijboer, Luca Braccioli, Ineke Slaper-Cortenbach, Annemieke Kavelaars, Frank van Bel, Cobi J Heijnen, Vanessa Donega, Cora H Nijboer, Luca Braccioli, Ineke Slaper-Cortenbach, Annemieke Kavelaars, Frank van Bel, Cobi J Heijnen

Abstract

Intranasal treatment with C57BL/6 MSCs reduces lesion volume and improves motor and cognitive behavior in the neonatal hypoxic-ischemic (HI) mouse model. In this study, we investigated the potential of human MSCs (hMSCs) to treat HI brain injury in the neonatal mouse. Assessing the regenerative capacity of hMSCs is crucial for translation of our knowledge to the clinic. We determined the neuroregenerative potential of hMSCs in vitro and in vivo by intranasal administration 10 d post-HI in neonatal mice. HI was induced in P9 mouse pups. 1×10(6) or 2×10(6) hMSCs were administered intranasally 10 d post-HI. Motor behavior and lesion volume were measured 28 d post-HI. The in vitro capacity of hMSCs to induce differentiation of mouse neural stem cell (mNSC) was determined using a transwell co-culture differentiation assay. To determine which chemotactic factors may play a role in mediating migration of MSCs to the lesion, we performed a PCR array on 84 chemotactic factors 10 days following sham-operation, and at 10 and 17 days post-HI. Our results show that 2×10(6) hMSCs decrease lesion volume, improve motor behavior, and reduce scar formation and microglia activity. Moreover, we demonstrate that the differentiation assay reflects the neuroregenerative potential of hMSCs in vivo, as hMSCs induce mNSCs to differentiate into neurons in vitro. We also provide evidence that the chemotactic factor CXCL10 may play an important role in hMSC migration to the lesion site. This is suggested by our finding that CXCL10 is significantly upregulated at 10 days following HI, but not at 17 days after HI, a time when MSCs no longer reach the lesion when given intranasally. The results described in this work also tempt us to contemplate hMSCs not only as a potential treatment option for neonatal encephalopathy, but also for a plethora of degenerative and traumatic injuries of the nervous system.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. In vitro proliferation of hMSCs.
Figure 1. In vitro proliferation of hMSCs.
Proliferating capacity of hMSCs in vitro. 1000 hMSCs were plated (T0) and proliferation was assessed at 4(T0), 24(T24), 48(T48) and 96(T96) hours after plating the MSCs by adding 3H-thymidine to the culture and measuring 3H-thymidine uptake 16 hours later. Data represent mean ± SEM. ** p<0.01; *** p<0.001 by ANOVA and Bonferroni post-hoc test. (n = 10 wells for each condition).
Figure 2. hMSCs induce differentiation of mouse…
Figure 2. hMSCs induce differentiation of mouse NSCs in vitro.
In vitro mNSC transwell differentiation assay in co-culture with hMSCs. mNSCs were fixed at 4(T0) and 96(T96) hours after co-culture with hMSCs and stained for (A) nestin (green), (B) Olig2 (red), (C) GFAP (green) and (D) βIII-Tubulin (red). Data represent mean ± SEM. *** p<0.001 by Unpaired two-tailed T-test. Scale bar  = 100 µm. (n = 4 wells per condition).
Figure 3. PKH-26 labeled hMSCs migrate to…
Figure 3. PKH-26 labeled hMSCs migrate to the lesion site.
1×106 hMSCs were labeled with PKH-26 and administered intranasally at 10 days after HI. (A+B) Mice were terminated 24 hours after hMSC treatment. (A) PKH-26+ hMSCs in the ipsilateral damaged cortex. (B) Contralateral cortex shows no PKH-26+ signal. (C) qPCR validation of PCR array confirmed the upregulation of five genes 10 days after HI. At 17 days after HI 2 genes were down-regulated in comparison to 10 days following HI. (HI n = 10; sham n = 6). (D) hMSCs express CXCR3, which increases after co-culture with HI brain extract (n = 2). Data represent mean ± S.E.M. *p<0.05; **p<0.01; ***p<0.001 by ANOVA and Bonferroni post-hoc test. Dashed line  =  lesion border; Asterisk  =  lesion site. Blue  =  Dapi staining. Scale bar  = 50 µm.
Figure 4. Dose effect of hMSC on…
Figure 4. Dose effect of hMSC on motor performance and lesion volume.
Mice were treated intranasally with either 1×106 or 2×106 hMSCs or vehicle at 10 days after HI. (A) Preference to use the unimpaired forepaw in the cylinder rearing test (CRT) was assessed at 28 days after HI. Sham-operated littermates (Sham) were used as controls. (B–C) Quantification of ipsilateral MAP2 (B) and MBP (C) area loss measured as 1- (ipsi-/contralateral MAP2- or MBP-positive area) at 28 days after HI. Representative sections of MAP2 (D) and MBP (E) staining. Data represent mean ± SEM. **p<0.01; ***p<0.001 by ANOVA and Bonferroni post-hoc test. Sham n = 13; Vehicle n = 21; 1×106 hMSC n = 11; 2×106 hMSC n = 12. Data presented in this figure are results from pups pooled out of 11 different litters. Treatment groups were randomly distributed between litters.
Figure 5. hMSCs reduce the activation of…
Figure 5. hMSCs reduce the activation of glial cells at 28 days after HI.
Mice were treated with either 1×106 or 2×106 hMSCs or vehicle intranasally at 10 days following HI. Mice were sacrificed 28 days after HI. (A) Schematic overview of fields quantified. (B) Quantification of Iba-1+ signal/mm2 or (C) GFAP+ signal/mm2. (D–G) Representative sections of Iba-1 (red) and GFAP (green) expression after sham-operation (D), vehicle (E), 1×106 hMSCs (F) or 2×106 hMSCs (G). Sections are counterstained with DAPI (blue). Scale bar  = 100 µm. Data represent mean ± SEM. * p<0.05; **p<0.01; ***p<0.001 by ANOVA and Bonferroni post-hoc test (Sham and Vehicle n = 4; 1×106 and 2×106 MSC n = 3).

References

    1. Dammann O, Ferriero D, Gressens P (2011) Neonatal encephalopathy or hypoxic ischemic encephalopathy? Appropriate terminology matters. Pediatr Res 70: 1–2.
    1. De Haan M, Wyatt JS, Roth S, Vargha-Khadem F, Gadian D, et al. (2006) Brain and cognitive-behavioural development after asphyxia at term birth. Dev Sci 9: 350–358.
    1. Ferriero DM (2004) Neonatal brain injury. N Engl J Med 351: 1985–1995.
    1. Graham EM, Ruis KA, Hartman AL, Northington FJ, Fox HE (2008) A systematic review of the role of intrapartum hypoxia-ischemia in the causation of neonatal encephalopathy. Am J Obstet Gynecol 199: 587–595.
    1. van Handel M, Swaab H, de Vries LS, Jongmans MJ (2007) Long-term cognitive and behavioural consequences of neonatal encephalopathy following perinatal asphyxia: a review. Eur J Pediatr 166: 645–654.
    1. Wang CC, Chen CH, Lin WW, Hsieh PCH, Lai PH, et al. (2008) Direct intramyocardial injection of mesenchymal stem cell sheet fragments improves cardiac functions after infarction. Cardiovasc Res 77: 515–524.
    1. Gonzalo-Daganzo R, Regidor C, Martin-Donaire T, Rico MA, Bautista G, et al. (2009) Results of a pilot study on the use of third –party donor mesenchymal stromal cells in cord blood transplantation in adults. Cytotherapy 11: 278–288.
    1. Deans RJ, Moseley AB (2000) Mesenchymal stem cells: Biology and potential clinical uses. Exp Hematol 28: 875–884.
    1. Zhang R, Liu Y, Yan K, Chen L, Chen XR, et al. (2013) Anti-inflammatory and immunomodulatory mechanisms of mesenchymal stem cell transplantation in experimental traumatic brain injury. J Neuroinflammation 10: 106–118.
    1. Salem HK, Thiemermann C (2010) Mesenchymal stromal cells: Current understanding and clinical status. Stem Cells 28: 585–596.
    1. Donega V, van Velthoven CT, Nijboer CH, van Bel F, Kas MJ, et al. (2013) Intranasal mesenchymal stem cell treatment for neonatal brain damage: Long-Term cognitive and sensorimotor improvement. PLoS ONE 8: e51253.
    1. Borlongan CV, Weiss MD (2011) Baby STEPS: A giant leap for cell therapy in neonatal brain injury. Pediatr Res 70: 3–9.
    1. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ (2010) Repeated mesenchymal stem cell treatment after neonatal hypoxia-ischemia has distinct effect on formation and maturation of new neurons and oligodendrocytes leading to restoration of damage, corticospinal motor tract activity, and sensorimotor function. J Neurosci 28: 9603–11.
    1. Titomanlio L, Kavelaars A, Dalous J, Mani S, El Ghouzzi V, et al. (2011) Stem cell therapy for neonatal brain injury: perspectives and challenges. Ann Neurol 70: 698–712.
    1. van Velthoven CT, van de Looij Y, Kavelaars A, Zijlstra J, van Bel F, et al. (2012) Mesenchymal stem cells restore cortical rewiring after neonatal ischemia in mice. Ann Neurol 71: 785–96.
    1. Velthoven C, Kavelaars A, van Bel F, Heijnen CJ (2011) Mesenchymal stem cell transplantation changes the gene expression profile of the neonatal ischemic brain. Brain Behav Immun 25: 1342–1348.
    1. Yasuhara T, Hara K, Maki M, Mays RW, Deans RJ, et al. (2008) Intravenous grafts recapitulate the neurorestoration afforded by intracerebrally delivered multipotent adult progenitor cells in neonatal hypoxic-ischemic rats. J Cereb Blood Flow Metab 28: 1804–1810.
    1. Tse WT, Pendlenton JD, Beyer WM, Egalka MC, Guinan EC (2003) Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation 75: 389–397.
    1. Toyoma K, Honmou O, Harada K, Suzuki J, Houkin K, et al. (2009) Therapeutic benefits of angiogenetic gene-modified human Mesenchymal stem cells after cerebral ischemia. Exp Neurol 216: 47–55.
    1. Liu H, Honmou O, Harada K, Nakamura K, Houkin K, et al. (2006) Neuroprotection by PIGF gene-modified human mesenchymal stem cells after cerebral ischemia. Brain 129: 2734–2745.
    1. Yang B, Xi XP, Aronowski J, Savitz SI (2012) Ischemic stroke may activate bone marrow mononuclear cells to enhance recovery after stroke. Stem Cells Dev 21: 3332–3340.
    1. Prins HJ, Rozemuller H, Vonk–Griffioen S, Verweij VG, Dhert WJ, et al. (2009) Bone forming capacity of mesenchymal stromal cells when cultured in presence of human platelet lysate as substitute for fetal calf serum. Tissue Eng Part A 15: 3741–3751.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marinin F, et al. (2006) Minimal criteria for defining multipotent Mesenchymal stromal cells. The international Society for Cellular Therapy. Cytotherapy 8: 315–317.
    1. Horwitz EM, Le BK, Dominici M, Mueller I, Slaper-Cortenbach I, et al. (2005) Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 7: 393–395.
    1. Van Praag H, Kempermann G, Gage FH (1999) Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 2: 266–270.
    1. Van Velthoven CT, Braccioli L, Willemen HL, Kavelaars A, Heijnen CJ (2014) Therapeutic potential of genetically modified mesenchymal stem cells after neonatal hypoxic-ischemic brain damage. Mol Ther 22: 645–654.
    1. Croft A, Przyborski SA (2009) Mesenchymal stem cells expressing neural antigens instruct a neurogenic cell fate on neural stem cells. Exp Neurol 216: 329–341.
    1. Kempermann G, Gast D, Kronenberg G, Yamagushi M, Gage FH (2003) Early determination and long-term persistence of adult-generated new neurons in the hippocampus of mice. Development 130: 391–399.
    1. Hack MA, Sugimori M, Lundberg C, Nakafuku M, Gotz M (2004) Regionalization and fate specification in neurospheres: the role of Olig2 and Pax6. Mol Cell Neurosci 25: 664–678.
    1. Hack MA, Saghatelyan A, de Chevigny A, Pfeifer A, Ashery-Padan R, et al. (2005) Neuronal fate determinants of adult olfactory bulb neurogenesis. Nat Neurosci 8: 865–872.
    1. Donega V, Nijboer CH, van Tilborg G, Dijkhuizen RM, Kavelaars A, et al. (2014) Intranasally administered mesencymal stem cells promote a regenerative niche for repair of neonatal ischemic brain injury. Exp Neurol 261: 53–64.
    1. Lee JA, Kim BI, Jo CH, Choi CW, Kim EK, et al. (2010) Mesenchymal stem cell transplantation for hypoxic-ischemic brain injury in neonatal rat model. Pediatr Res 67: 42–46.
    1. Silver J, Miller JH (2004) Regeneration beyond the glial scar. Nat Rev Neurosci 5: 146–156.
    1. Sofroniew MV (2014) Multiple Roles for astrocytes as effectors of cytokines and inflammatory mediators. Neuroscientist 20: 160–72.
    1. Wake H, Moorhouse AJ, Miyamoto A, Nabekura J (2013) Microglia: actively surveying and shaping neuronal circuit structure and function. Trends Neurosci 36: 209–217.
    1. Donega V, van Velthoven CT, Nijboer CH, Kavelaars A, Heijnen CH (2013) The endogenous regenerative capacity of the newborn brain: Boosting neurogenesis with mesenchymal stem cell treatment. JCBFM 33: 625–634.
    1. Ziebell JM, Morganti-Kossmann MC (2010) Involvement of pro-and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury. Neurotherapeutics 7: 22–30.
    1. Reglero-Real N, Marcos-Ramiro B, Millan J (2012) Endothelial membrane reorganization during leukocyte extravasation. Cell Mol Life Sci 69: 3079–3099.
    1. Ransohoff RM, Hamilton TA, Tani M, Stoler MH, Shick HE, et al. (1993) Astrocyte expression of mRNA encoding cyotkines IP-10 and JE/MCP-1 in experimental autoimmune encephalomyelitis. FASEB J 7: 592.
    1. Ubogu EE, Cossoy MB, Ransohoff RM (2006) The expression and function of chemokines involved in CNS inflammation. Trends in Pharmacol Sci 27: 48–55.

Source: PubMed

3
Prenumerera