Elsevier

Neurobiology of Aging

Volume 51, March 2017, Pages 83-96
Neurobiology of Aging

Regular article
Administrations of human adult ischemia-tolerant mesenchymal stem cells and factors reduce amyloid beta pathology in a mouse model of Alzheimer's disease

https://doi.org/10.1016/j.neurobiolaging.2016.11.009Get rights and content

Abstract

The impact of human adult ischemia-tolerant mesenchymal stem cells (hMSCs) and factors (stem cell factors) on cerebral amyloid beta (Aβ) pathology was investigated in a mouse model of Alzheimer's disease (AD). To this end, hMSCs were administered intravenously to APPPS1 transgenic mice that normally develop cerebral Aβ. Quantitative reverse transcriptase polymerase chain reaction biodistribution revealed that intravenously delivered hMSCs were readily detected in APPPS1 brains 1 hour following administration, and dropped to negligible levels after 1 week. Notably, intravenously injected hMSCs that migrated to the brain region were localized in the cerebrovasculature, but they also could be observed in the brain parenchyma particularly in the hippocampus, as revealed by immunohistochemistry. A single hMSC injection markedly reduced soluble cerebral Aβ levels in APPPS1 mice after 1 week, although increasing several Aβ-degrading enzymes and modulating a panel of cerebral cytokines, suggesting an amyloid-degrading and anti-inflammatory impact of hMSCs. Furthermore, 10 weeks of hMSC treatment significantly reduced cerebral Aβ plaques and neuroinflammation in APPPS1 mice, without increasing cerebral amyloid angiopathy or microhemorrhages. Notably, a repeated intranasal delivery of soluble factors secreted by hMSCs in culture, in the absence of intravenous hMSC injection, was also sufficient to diminish cerebral amyloidosis in the mice. In conclusion, this preclinical study strongly underlines that cerebral amyloidosis is amenable to therapeutic intervention based on peripheral applications of hMSC or hMSC factors, paving the way for a novel therapy for Aβ amyloidosis and associated pathologies observed in AD.

Introduction

Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia in the Western world. AD is neuropathologically characterized by the formation of extracellular senile plaques, primarily composed of polymerized amyloid beta (Aβ) peptides and the intracellular deposition of hyperphosphorylated tau proteins into neurofibrillary tangles (Selkoe, 1991). According to the amyloid cascade hypothesis, the extracellular deposition of Aβ is a critical and central event in the disease process leading to the formation of neurofibrillary tangles, neuroinflammation, cell death, and dementia (Hardy and Allsop, 1991). There is no cure on the horizon for this devastating disease that represents one of today's major healthcare challenges due to its severe socio-economic burden (Brookmeyer et al., 2007, Waite, 2015).

Human adult ischemia-tolerant mesenchymal stem cells (hMSCs) constitute a promising therapeutic approach for the treatment of various neurodegenerative disorders including AD (Fan et al., 2014). However, food and drug administration-approved clinical trials currently evaluating the impact of hMSC on AD are marginal. Therefore, an hMSC-based AD therapy represents a large untapped potential waiting to be exploited. Several preclinical research laboratories have reported a beneficial effect of mesenchymal stem cells on cerebral amyloidosis, adult neurogenesis, or memory impairments in models of AD (e.g., Salem et al., 2014, Yan et al., 2014, Yun et al., 2013). However, the majority of these therapeutic initiatives rely on the direct stem cell delivery into the brain through intracerebral or intracerebroventicular injection. These routes of administration entail a major hurdle for clinical applications due to their invasiveness and possible complications (Reyes et al., 2015). This difficulty has certainly hampered to a large extent the clinical translation of these preclinical findings. In contrast, intravenous delivery is fast, easy and complications are rarely observed. To date, only a limited number of preclinical studies have evaluated the impact of intravenous hMSC injections on cerebral amyloidosis (e.g., Kim et al., 2013).

In this present study, we investigated the impact of hMSC and mesenchymal stem cell factors (SCF) on cerebral Aβ pathology in a mouse model of AD. The hMSCs used in this study are bone marrow derived, ischemia-tolerant, cultured under a controlled, low physiological level of oxygen (5%), and manufactured under clinical good manufacturing process conditions. Importantly, these hMSC express negligible levels of human leukocyte antigen-D related (HLA-DR) cell surface receptor and have higher migratory ability as compared to cells cultured at the normal oxygen level (Vertelov et al., 2013). Quantitative reverse transcriptase polymerase chain reaction (RT-PCR) biodistribution revealed that intravenously delivered hMSC are detected in the brain at 1 hour after delivery, decreasing after 1 day, and subsequently dropping below detection level at 1 week after injection. Brain immunohistochemistry demonstrated that intravenously injected hMSCs could be detected in both the cerebrovasculature and in the brain parenchyma. A single intravenous hMSC injection was effective in reducing soluble cerebral Aβ levels at 1 week after delivery in both early and late stage of plaque development. The levels of several major Aβ-degrading enzymes were significantly increased in hMSC-treated mice. In addition, the levels of several proinflammatory cytokines such as Tumor Necrosis Factor alpha (TNFα) were decreased at 1 week after hMSC injection. As a complement to single hMSC delivery, the impact of a repeated intravenous hMSC delivery was investigated on cerebral amyloidosis in APPPS1 mice. Repeated delivery of hMSC (1 injection/wk for 10 weeks) safely reduced cerebral Aβ plaques in both young and aged APPPS1 animals analyzed 1 week after the last injection. Concomitantly, microglial coverage was diminished in hMSC-treated APPPS1 mice. No increase of vascular amyloid or manifestation of microhemorrhages was observed following the repeated intravenous hMSC delivery. Last but not least, in the absence of hMSC injection, proteins secreted by hMSC in culture could also significantly reduce cerebral amyloidosis in APPPS1 mice following repeated intranasal applications for a total of 3 weeks.

In summary, our preclinical results demonstrate that single and repeated intravenous administration of hMSC safely reduces Aβ pathology in the APPPS1 model of AD. The successful reduction of cerebral amyloidosis following intranasal application of hMSC factors invites the next step, that is, clinical studies using these factors as complementary treatment to stem cell injections.

Section snippets

Animals and experimental study design for in vivo experiments

For the in vivo experiments, APPPS1 transgenic mice were obtained from M. Jucker (HIH, Tübingen, Germany) and maintained at the EPFL animal core facility. The mice coexpress under the control of the Thy-1 promoter the KM670/671NL Swedish mutation of human amyloid precursor protein (APP) and the L166P mutation of human presenilin 1 (PS1) and usually show the first amyloid plaques in the cortex at an age of 6–8 weeks. No gender effects in Abeta levels and amyloid deposition are observed in this

Reduction of cerebral soluble Aβ levels in APPPS1 mice following a single injection of hMSC

The Alzheimer model used in this study is the APPPS1 mouse which co-expresses KM670/671NL mutated APP and L166P mutated PS1 under the control of a neuron-specific Thy1 promoter. Cerebral amyloidosis starts in these mice as early as 6–8 weeks of age, and the number of Aβ plaques increases steadily. Single intravenous injections of hMSC were performed in APPPS1 mice (Table 1), and the levels of Aβ42 were analyzed by enzyme-linked immunosorbent assay 1 week after delivery. APPPS1 animals at both

Discussion

In the present work, we have first investigated the impact of a single intravenous hMSC injection in a mouse model of AD and demonstrated that hMSC delivery reduced soluble cerebral Aβ in both young and aged APPPS1 mice at 1 week after delivery. In addition, the levels of several Aβ degrading enzymes were significantly decreased. These results suggest a beneficial impact of hMSC through upregulation of the levels of certain Aβ-degrading enzymes leading to increased Aβ clearance. Analysis of

Conclusions

Adult ischemia-tolerant mesenchymal stem cells (hMSCs) represent a promising yet largely unexploited therapeutic approach for AD. Using a mouse model of cerebral Aβ amyloidosis, we have demonstrated that single and repeated intravenous hMSC injections reduce cerebral Aβ pathology and neuroinflammation with no increase in cerebral amyloid angiopathy or microhemorrhages. Observations of changes in Aβ degrading enzymes and cytokine expression suggest possible mechanisms underlying the actions of

Disclosure statement

The authors declare having competing financial interests. Dr Bolmont, Dr Lukashev, and Cheatham acknowledge their employment at Stemedica International as Chief Scientist, General Manager, and Business Manager, respectively.

Acknowledgements

This work was supported by a grant from the Swiss Commission for Technology and Innovation (BT, PFLS-LS 13964.1). The authors thank Prof Jucker M. (HIH, Tübingen) for providing the APPPS1 animals as well as for insightful comments on the manuscript. The authors thank Dr Yuri Kudinov (StemCutis) for his technical assistance. LA, LT, and BT designed research; HT and ZV performed biodistribution studies; DN and MC performed immunohistological experiments; LY performed quantification analyses; and

References (37)

  • J. Apelt et al.

    Aging-related down-regulation of neprilysin, a putative beta-amyloid-degrading enzyme, in transgenic Tg2576 Alzheimer-like mouse brain is accompanied by an astroglial upregulation in the vicinity of beta-amyloid plaques

    Neurosci. Lett.

    (2003)
  • M. Bacher et al.

    The role of macrophage migration inhibitory factor in Alzheimer's disease

    Mol. Med.

    (2010)
  • F. Balkwill et al.

    Cancer: an inflammatory link

    Nature

    (2004)
  • W.A. Banks et al.

    Passage of amyloid beta protein antibody across the blood-brain barrier in a mouse model of Alzheimer's disease

    Peptides

    (2002)
  • R.J. Boado et al.

    Pharmacokinetics and brain uptake in the rhesus monkey of a fusion protein of arylsulfatase a and a monoclonal antibody against the human insulin receptor

    Biotechnol. Bioeng.

    (2013)
  • R. Brookmeyer et al.

    Forecasting the global burden of Alzheimer's disease

    Alzheimers Dement.

    (2007)
  • S. Bürger et al.

    Vascular endothelial growth factor (VEGF) affects processing of amyloid precursor protein and beta-amyloidogenesis in brain slice cultures derived from transgenic Tg2576 mouse brain

    Int. J. Dev. Neurosci.

    (2009)
  • J. Chen et al.

    Intravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in rats

    Circ. Res.

    (2003)
  • L.M. Coussens et al.

    Inflammation and cancer

    Nature

    (2002)
  • X. Fan et al.

    Stem-cell challenges in the treatment of Alzheimer's disease: a long way from bench to bedside

    Med. Res. Rev.

    (2014)
  • W. Farris et al.

    Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • C. Gallina et al.

    A new paradigm in cardiac regeneration: the mesenchymal stem cell secretome

    Stem Cells Int.

    (2015)
  • M. Guillot-Sestier et al.

    Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology

    Neuron

    (2015)
  • J. Hardy et al.

    Amyloid deposition as the central event in the aetiology of Alzheimer's disease

    Trends Pharmacol. Sci.

    (1991)
  • M. Hernández-Guillamon et al.

    Neuronal TIMP-1 release accompanies astrocytic MMP-9 secretion and enhances astrocyte proliferation induced by beta-amyloid 25-35 fragment

    J. Neurosci. Res.

    (2009)
  • K.K. Hirschi et al.

    Gap junction communication mediates transforming growth factor-beta activation and endothelial-induced mural cell differentiation

    Circ. Res.

    (2003)
  • P. Johansson et al.

    Serum but not cerebrospinal fluid levels of insulin-like growth factor-I (IGF-I) and IGF-binding protein-3 (IGFBP-3) are increased in Alzheimer's disease

    Psychoneuroendocrinology

    (2013)
  • K.S. Kim et al.

    Long-term immunomodulatory effect of amniotic stem cells in an Alzheimer's disease model

    Neurobiol. Aging

    (2013)
  • Cited by (0)

    View full text