Skip to main content

Advances in intranasal application of stem cells in the treatment of central nervous system diseases

Abstract

Stem cells are characterized by their self-renewal and multipotency and have great potential in the therapy of various disorders. However, the blood–brain barrier (BBB) limits the application of stem cells in the therapy of neurological disorders, especially in a noninvasive way. It has been shown that small molecular substances, macromolecular proteins, and even stem cells can bypass the BBB and reach the brain parenchyma following intranasal administration. Here, we review the possible brain-entry routes of transnasal treatment, the cell types, and diseases involved in intranasal stem cell therapy, and discuss its advantages and disadvantages in the treatment of central nervous system diseases, to provide a reference for the application of intranasal stem cell therapy.

Background

Central nervous system (CNS) diseases, such as cerebrovascular diseases, Alzheimer’s disease (AD), and Parkinson’s disease (PD), mainly involve the injury, degeneration, and loss of neural cells. Stem cells are a type of cells with the potential of self-renewal and multipotency, which exhibit great potential in replacing damaged neural cells and improving neurological function [1]. Stem cell therapy has great clinical prospects in the treatment of CNS diseases, with several ongoing clinical trials [2].

The blood-brain barrier (BBB) is an obstacle for delivery of most drugs to the CNS via traditional treatment routes (such as intravenous injection) that reduces therapeutic efficacy of most drugs. Development of compounds that can bypass the BBB is always a challenge for drug development. Stem cell transplantation has therapeutic potential in various diseases such as PD and AD. However, stem cells therapy in the brain always requires an invasive approach, such as stereotactic or intrathecal injection. The invasive approach limits the application of stem cell transplantation in the therapy of neurological disorders. Many studies have observed that intranasal application (INA) can bypass the BBB and enable the delivery of small molecular substances and macromolecular proteins to the CNS [3, 4]. Moreover, compared with stereotactic brain injection and intrathecal injection, INA is less invasive and can be done repeatedly. Intranasal delivery of drugs to the brain was first established by William Frey in 1989 [5]. Since then, a lot of other substances such as chemicals [6], plasmids [7], peptides [8, 9], viruses [10], and metals [11] have been delivered successfully through the intranasal cavity. In 2009, Danielyan et al. firstly delivered fluorescently labeled rat mesenchymal stem cells (MSCs) or human glioma cells to rodent brains via the nasal cavity. The intranasally delivered cells were observed in the olfactory bulb, cortex, cerebellum, and subarachnoid space [5]. Since then, several studies have shown that stem cells derived from different tissues can also be delivered to the CNS by INA, exhibiting therapeutic effects [12,13,14,15]. The intranasally delivered cells can be observed in the olfactory bulb, cortex, cerebellum, and subarachnoid space [12]. The intranasally delivered cells even exhibit more extensive migration to the injured or damaged areas in murine models of hypoxia-ischemia and ischemic stroke [16, 17], or brain tumors [18, 19]. In contrast, they seem not to localize to the subventricular zone; the region where endogenous adult stem cells are enriched [17]. However, it is worth noting that intranasally delivered stem cells show a low migration efficiency since a large number of cells remained in the upper nasal cavity 1 h after intranasal administration [12, 14]. Here, we review the possible brain-entry routes and the types of stem cells in the intranasal treatment of CNS diseases. Due to limited space, our current review only discusses INA of stem cells, rather than other substances that can be delivered via INA, which are reviewed elsewhere [20,21,22,23,24,25].

Possible brain-entry routes of INA

Since 1989, Frey proved the existence of the route from the nose to the brain and opened a new era for INA [5]. In INA, substances deposit in the epithelial cell layer of the nasal cavity and reach the brain parenchyma either directly by migrating along the olfactory or trigeminal nerve, or through the cerebrospinal fluid circulation [24]. Once the substances reach their specific location, they can be transported to the CNS through the intracellular and paracellular transport pathways (Fig. 1).

Fig. 1
figure 1

Possible brain-entry routes of substances by INA (in rodents as an example). After intranasal injection into the nasal cavity of rodents, drugs or cells are transported via the intracellular and/or paracellular pathways, and pass through the olfactory epithelium and reach the olfactory bulb along the olfactory nerve, or pass through the respiratory epithelium to reach the olfactory bulb/brainstem along the ophthalmic or maxillary branches of the trigeminal nerve. After reaching the CNS, drugs or cells are further distributed in the brain through the cerebrospinal fluid and/or perivascular spaces

Intracellular transport pathway

Receptor-mediated endocytosis is the main mechanism by which substances enter the CNS through the intracellular transport pathway. Thorne et al. found that horseradish- peroxidase-conjugated wheat germ agglutinin (WGA-HRP) could reach the brain parenchyma via receptor-mediated endocytosis of the olfactory receptor neurons (ORNs) [26]. After being internalized by ORNs, the substances form vesicles and move along the axons. Once the vesicles reach the nerve endings, the substances are released by exocytosis to reach the periglomerular cell layer of the olfactory bulb. There are also endocytosis-mediating receptors on the surface of mitral cells within the periglomerular cell layer, which produce secondary transport of the substances. Through the above process, substances can diffuse to multiple brain regions, mainly including the olfactory nodule, piriform plexus, amygdala, hypothalamus, and inner olfactory plexus [27]. In addition to the ORNs, Robertson et al. [28] and Anton et al. [29] confirmed that WGA-HRP enters the CNS through neuroendocytosis of the ophthalmic and maxillary branches of the trigeminal nerve.

Distribution of substances in the CNS can be observed within a few hours after intranasal injection. In contrast, it takes longer for substances to enter the CNS via the intracellular transport pathway. In addition, the characteristics, molecular weight of the substances, and the selectivity of receptors limit the transportation efficiency [21], suggesting that intracellular transport may not be the main route after INA. Further investigations are required to validate whether stem cells enter the brain through the intracellular transport pathway.

Paracellular transport pathway

In physiological structures, cells connect with one another through different structures, such as the common tight junctions [30]. These connections are innately impermeable to macromolecular drugs. However, in the nasal cavity, they are permeable due to the continuous renewal of the neurons and basal cells, which provides the basis for paracellular transport. Through the paracellular transport pathway, drugs can reach the olfactory bulb and other areas of the CNS within a few to 30 min [31].

Perineural pathway

In the nervous system, oligodendrocytes in the CNS and Schwann cells in the peripheral nervous system form myelin sheaths, which surround the axons or the long dendrites of sensory neurons. The space between the myelin sheaths and neurons is known as the perineuronal space. Myelin sheaths have plasticity and play an important role in nerve signal transduction. Additionally, they support and guide neuron migration and participate in substance metabolism and nutritive function [32]. They also form the physiological basis for substances to enter the CNS from the olfactory epithelium along the olfactory nerve or from the respiratory epithelium along the trigeminal nerve. The paracellular pathway of drugs delivered intranasally starts from the paracellular fissure of olfactory epithelial cells (Fig. 1). The tight junctions between the olfactory neurons and the supporting cells enable substances to move from the nasal cavity to the lamina propria [33]. Most of the substances are absorbed from the lamina propria by local blood vessels or lymphatic vessels and enter the cervical lymph nodes. The remaining substances are transported through the perineural space, along the olfactory nerve, and terminate at the olfactory bulb. Substances can also move along the trigeminal nerve, pass through the cribriform plate, and reach the olfactory bulb or brain stem [34]. A previous study showed that MSCs could pass through the cribriform plate and be detected in the olfactory nerve layer of the olfactory bulb 2 h after nasal administration [14], but the mechanism remains unclear.

Perivascular pathway

Perivascular spaces play an important role in the transport of substances to deep parts of the brain after arriving in the CNS via the perineural pathway, especially in the process of rapid distribution of small molecules in brain parenchyma at the early stage [35]. Perivascular spaces are formed between the endothelium of large and medium-sized arteries and veins in the brain and the glial cells surrounding them. In research on the brain lymphatic system, Iliff et al. suggested that most cerebrospinal fluid from the subarachnoid space flows back to brain parenchyma through perivascular spaces and exchanges with brain tissue fluid [36]. A similar distribution was observed between tracers injected intranasally and via the subarachnoid space, suggesting that perivascular spaces participate in the transport route after INA [37, 38]. This raises the question whether stem cells could distribute in the brain through this pathway. Although the structure of perivascular spaces suggests that it is difficult for macromolecular substances to exist in perivascular spaces [31], some results have shown that colocalization of MSC markers and the vascular endothelial marker von Willebrand factor is observed 7 days after injection of MSCs [14]. Endogenous stem cells can migrate from the subventricular zone (SVZ) to the olfactory bulb through rostral migration using perivascular spaces as a scaffold [39]. Whether INA of exogenous stem cells can take advantage of this pathway is a question worthy of further investigation.

Types of stem cells administered by transnasal route

No study has compared the function of all types of stem cells at the same time. Some in vivo investigations have shown that the fusogenic role of microglia could be even more important after neural stem cell transplantation into brains affected by neurodegenerative diseases associated with microglial activation. Among cultured stem cells in vitro, it has been clearly shown that embryonic stem cells readily fuse with microglia. These microglia then, in turn, go on to fuse with mature neurons [20]. However, definitively demonstrating which type of intranasally delivered stem cells perform best in reaching appropriate targets and have the best restorative or therapeutic effect may be difficult. We agree with the conclusion of Danielyan et al. that the motility and migratory potential of stem cells can serve as predictors of their therapeutic efficacy because they all relate to the functional heterogeneity of stem cells [40].

Neural stem cells (NSCs)

NSCs have the ability to differentiate into neurons and glia, thus harboring advantages in the treatment of neurological diseases. NSCs can be obtained from either isolating the cells from the SVZ of newborn or E14 rats [41] or from differentiation of other pluripotent stem cells such as hematopoietic stem cells, embryonic stem cells, MSCs, and induced pluripotent stem cells (iPSCs) [42]. INA of NSCs has been used for treatment of a variety of neurological diseases. For example, INA of NSCs, combined with other therapy, increases the survival in a murine model of glioblastoma of the brain (GBM) [19, 43, 44]. Intranasal injection of NSCs has also been used in the treatment of brain injury [45] and PD [46]. However, in terms of clinical application, as the main source of human NSCs is fetal brain tissue, its clinical application is limited by ethical issues [13].

MSCs

MSCs are adult stem cells derived from various sources including the bone marrow, fat, umbilical cord, and cord blood. Bone marrow-derived MSCs (BM-MSCs) contain a subgroup expressing nestin, a marker of NSCs, and can differentiate into glial cells and neurons with complete functions, promoting nerve tissue regeneration [47]. Therefore, INA of BM-MSCs has been widely used in research into treating brain injury [48], neurodegenerative diseases, and glioma [15]. Several clinical trials are ongoing related to therapy of CNS diseases by intranasal injection of MSCs.

In addition to BM-MSCs, some recent studies have explored the therapeutic potential of intranasal injection of dental pulp stem cells (DPSCs) in treating PD [49]. DPSCs are neural-crest-derived ectodermal MSCs with strong self-renewal and multipotent capacity. They can be isolated from extracted dental pulp easily and show great potential in the treatment of CNS diseases and retinopathy. Endometrial MSCs are also used in PD therapy [50].

iPSCs

iPSCs are a special type of stem cells that are engineered from somatic cells, and their properties resemble inner mass cells of the embryo. iPSCs are generated from the somatic cells through genetic engineering, by which somatic cells are reprogrammed to cells with pluripotent capacity similar to that of embryonic stem cells. This technology was first introduced by the Japanese scientist Yamanaka in 2006 [51]. The induction of NSCs or other nerve cells from autologous iPSCs reduces the immunogenicity of iPSCs, thus immune rejection after transplantation, and resolves the ethical issues of transplanting NSCs. Many studies have shown that INA of iPSCs can inhibit acute or chronic airway inflammation [52,53,54], but studies of INA of iPSCs or iPSC-derived cells in neurological diseases is still lacking.

INA of stem cells for therapy of CNS disorders

No study has shown that there are particular diseases that would benefit most from INA compared with other administration routes. Obviously, it is difficult to compare all diseases and all administration routes in the same study. Although INA it has no disease specificity, it also shows the advantages of its wide application in a variety of neurological diseases. INA offers advantages over intravenous or other routes of administration in several circumstances [13]: (1) it has a higher degree of delivery to the CNS; (2) it avoids first-pass metabolism; (3) it is noninvasive and easy to be administered, which allows repetitive administration if necessary; and (4) its adverse effects can be minimized since other healthy organs are not exposed to the therapeutic compound. These properties make intravenous administration one of the prime strategies for therapy of long-term neurological disorders with multiple pathological regions, such as AD and PD.

Neuroinflammation is an important factor in the occurrence and development of neurological diseases, especially in PD and AD. However, only drug treatment for neuroinflammation in neurological diseases is very hard to achieve the best. Fortunately, an increasing number of studies have confirmed that stem cells transplanted into the brain can regulate the microenvironment of lesions and alter inflammatory mediators, so as to reduce the symptoms of neurological diseases. Some studies have suggested that INA of stem cells can also play an anti-inflammatory role [55]. For example, Danielyan L et al. [56] reported noninvasive INA of MSCs to the brains of unilaterally 6-hydroxydopamine (6-OHDA)-lesioned rats. After 40–110 days of INA of stem cells, MSCs decreased the concentration of inflammatory cytokines interleukin (IL)-1β, IL-2, IL-6, IL-12, tumor necrosis factor, interferon-γ, and granulocyte–macrophage colony-stimulating factor in the lesioned side to their levels in the intact hemisphere. McDonald et al. [57] found that INA of a clinically relevant dose of human umbilical-cord-derived MSCs in a rat model of neonatal hypoxic–ischemic brain injury was neuroprotective by restoring neuronal cell numbers and reducing brain inflammation.

Neurodegenerative diseases

The common neurodegenerative diseases include PD, AD, Huntington’s disease, and amyotrophic lateral sclerosis, which are characterized by neuron degeneration and loss in the brain and/or spinal cord. In the treatment of neurodegenerative diseases, stem cells can replace degenerative cells or protecting cells via transferring neurotrophic factors [58] that exhibit therapeutic potential in preclinical research and ongoing clinical trials.

PD

The pathological features of PD are the loss of dopaminergic neurons in the substantia nigra and formation of Lewy bodies. Current treatments for PD mainly rely on drug therapy and auxiliary surgical treatment, which exhibits symptomatic attenuation in motor fluctuation [59]. The exploration of other therapeutic approaches for PD have shown promising results, such as stem cell therapy, immune and inflammatory therapy using antibodies, and gene therapy [60]. Stem cell transplantation prevents the loss of dopaminergic neurons the substantia nigra of PD patients. The main transplantation method is injecting the cells directly to the brain parenchyma via stereotactic injection, which is invasive. Thus, noninvasive INA has attracted much attention. Table 1 lists the research of intranasal stem cell treatment of PD in rodent models in recent years. In 2011, Danielyan et al. first attempted to treat PD with intranasal MSCs in a rat model with unilateral substantia nigra damage induced by 6-OHDA [56]. The MSCs entered, survived, and proliferated in brain parenchyma for at least 4.5 months. Intranasal transplantation of MSCs effectively improved the tyrosine hydroxylase (TH) level in the damaged substantia nigra and striatum, and the motor symptoms in the model rats [56]. Subsequently, BM-MSCs, NSCs, DPSCs, and endometrial MSCs were administered intranasally to treat PD model rodents. And all of them produced some therapeutic effects [46, 49, 50, 62, 63]. However, the entry pathway, influencing factors and the different effect of different type of intranasally injected stem cells, have not been clarified and are worthy of further exploration.

Table 1 Recent studies on the treatment of PD model by INA of stem cells

AD

AD is the most common neurodegenerative disease, which is pathologically characterized by accumulation of amyloid plaques formed by amyloid-β (Aβ) and neurofibrillary tangles consisting of hyperphosphorylated tau. Impaired synaptic plasticity and neurodegeneration have been observed in the brains of AD patients. Exogenous stem cells play an important role in the reconstruction and regeneration of failed neuronal circuits [64]. In 2014, the research group of Danielyan L utilized intranasal injection of BM-MSCs to treat AD transgenic APP/PS1 mice. On 7 days after INA, stem cells were found near the Aβ deposition in the hippocampus and occipital cortex. The transplanted cells were also Aβ positive, suggesting that they had phagocytic and scavenging effects on Aβ [62]. In 2015, Mita et al. found that after intranasal injection of the serum-free conditioned medium of stem cells derived from human exfoliated deciduous teeth (SHED-CM), the cognitive function of AD model mice was improved [65]. Intranasal injection of the soluble molecules [66] or exosomes [67] of MSCs ameliorates the inflammation of AD mice and reduces Aβ. Table 2 summarizes recent research of INA of stem cells in the treatment of AD model mice. Intranasal injection of MSCs improves the abnormal dopamine system and inflammatory response in Huntington’s disease [68].

Table 2 Recent studies on treatment of AD murine models by INA of stem cells

Cerebrovascular diseases

Stem cell therapy also has advantages in the prognosis and functional repair of cerebrovascular diseases, especially in some ischemic vascular diseases. In vivo, peripheral vascular endothelial progenitor cells can repair damaged vascular endothelial cells and promote vascular regeneration [69]. In 2012, Wei et al. investigated intranasal transplantation of BM-MSCs in a murine model of ischemic stroke. The stem cells reached the ischemic cortex and were deposited outside the blood vessels at 1.5 h after administration. The transplanted stem cells homed to the infarcted area, protecting the cells in it [16]. The conditioned medium of nasal MSCs also improves the symptoms of middle cerebral artery occlusion MCAO model rats and enhance vascular remodeling [70]. INA of stem cells also exerts therapeutic effects in model rodents with subarachnoid hemorrhage [71] or neonatal brain injury [72,73,74].

GBM

GBM is one of the most aggressive malignant tumors and has poor prognosis. Stereotactic injection of stem cells has produced encouraging results in the treatment of GBM. The poor repeatability and invasiveness of drug delivery limits its application. INA provides a good method of stem cell administration [15]. In 2012, Reitz et al. found that NSCs were detected around GBM tissues 6 h after INA, which reached a peak at 24 h and persisted in the brain for more than 5 days [44]. In 2016, Dey et al. significantly enhanced the ability of NSCs to target tumors by hypoxic preconditioning or overexpression of chemokine CXC receptor 4. The modified NSCs more effectively delivered oncolytic viruses to glioma and prolonged the survival time of experimental animals, in combination with radiotherapy [43]. Current studies suggest that intranasal stem cell therapy (especially NSCs), after combination with other treatments, has a good effect on the treatment and prognosis of GBM and has great prospects.

Limitations of intranasal stem cell therapy

Cell migration in the brain after INA

Fluorescent tracer technology can be used to monitor the distribution of cells after they enter the brain. As the first pioneers, Danielyan et al. intranasally delivered fluorescently labeled rat MSCs or human glioma cells to naïve mice and rats. The fluorescently labeled cells were found in the olfactory bulb, cerebellum, subarachnoid space, and cortex [56]. In another study by the same group, MSCs were distributed near Aβ in AD model mice [62]. Reitz et al. intranasally injected enhanced green fluorescent protein (eGFP)-labeled NSCs into a glioma mouse model and found the cells mainly accumulated near the tumor [44]. In our study to compare the biodistribution of human neural stem cell (hNSC) migration into the brain via INA, we found that hNSCs delivered by INA could migrate into the striatum, hippocampus, cerebellum, and spinal cord in MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) treated mice, but they could not migrate into CNS after INA in normal mice [75]. An increasing number of studies have suggested that biodistribution differs among brain regions over time due differences in disease models and delivery and quantity of stem cells. There is no clear answer as to which method is best to label or trace stem cells. Some scholars believe that in vivo neuroimaging may be good [19, 76]. However, from the results of our previous studies [75], in vivo tracing by positron emission tomography/computed tomography is not satisfactory, and more study is needed. A few tracking studies have shown the enter path of stem cells. For instance, Galeano et al. have labeled hMSCs with bright quantum dots in order to track the cells efficiently [14]. The use of stem cell exosomes in nasal drops to treat neurological disease has also received considerable attention. In 2019, Perets et al. confirmed that the exosomes of MSCs administered intranasally selectively target brain tissues in different models of neurological diseases [67]. This indicates that some secretory substances play an important role in the migration and homing of intranasally injected stem cells.

Efficiency and quantity of stem cell entry

Not all stem cells reach the brain parenchyma after INA. Carlos et al. showed that among the intranasally injected MSCs, only a small number entered the CNS through the cribriform plate, while most fell into the gap of the turbinate bones [14]. Danielyan et al. have used quantitative polymerase chain reaction analysis of eGFP DNA to show the yield of eGFP-MSCs after INA [56, 62]. They used 1*106 MSCs by INA, but only 1*104 were found in the olfactory bulb, 100 in the hippocampus and 10 in the cortex. Therefore, further research is needed to improve the efficiency of cell delivery. Some experiments have shown that hyaluronidase can greatly the permeability of olfactory epithelium and improve material utilization rate. This method has also been used in intranasal stem therapy [12, 15]. It should be noted that different concentrations of stem cells were used across the studies. Thus, does the initial cell concentration affect experimental results? A study on PD reported that the behavioral improvement in the low stem cell concentration treatment group was better than that of the high concentration group [50], but this result needs validation.

Body weight and age of model animals

The distribution of drugs or cells after intranasal injection exhibits variability in distinct animal models. Even in the same animal model, intranasal injection shows different delivery efficacy according to animal age and weight. Krishnan et al. injected the drug radiolabeled pralidoxime into the nasal cavity of model animals, which showed that compared with younger animals with lower body weight, older animals with higher body weight required a higher dose to reach the same drug concentration in the whole brain [77]. This suggested that the distribution of intranasally administered stem cells might be affected by the age and body weight of model animals.

Conclusions: summary and prospects

INA has the advantages of noninvasiveness and high practicability. It facilitates all kinds of drug molecules and stem cells to bypass the BBB and reach the CNS, which provides a noninvasive and effective method for stem cell therapy in CNS diseases. However, unlike small molecular substances, the transnasal transport routes of stem cells remain unclear. Optimization of approaches in improving the delivery and therapeutic efficiency and safety of intranasal transplantation of stem cells is required in future studies. There are still many issues to be considered in the translation from animal experiments to clinical trials (Table 3).

Table 3 Challenges of stem cells translation from animal experiments to clinical application via INA

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

Aβ:

Amyloid-β

AD:

Alzheimer’s disease

BBB:

Blood–brain barrier

BM-MSCs:

Bone-marrow-derived MSCs

CNS:

Central nervous system

DPSCs:

Dental pulp stem cells

hNSCs:

Human neural stem cells

INA:

Intranasal application

iPSCs:

Induced pluripotent stem cells

MSCs:

Mesenchymal stem cells

NSCs:

Neural stem cells

ORNs:

Olfactory receptor neurons

PD:

Parkinson’s disease

SVZ:

Subventricular zone

TH:

Tyrosine hydroxylase

vWF:

von Willebrand factor

WGA-HRP:

Wheat Germ Agglutinin

References

  1. Bhagavati S. Stem cell therapy: challenges ahead. Indian J Pediatr. 2015;82(3):286–91. https://doi.org/10.1007/s12098-014-1521-5.

    Article  PubMed  Google Scholar 

  2. Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell. 2015;17(1):11–22. https://doi.org/10.1016/j.stem.2015.06.007.

    Article  CAS  PubMed  Google Scholar 

  3. Li YH, Feng L, Zhang GX, Ma CG. Intranasal delivery of stem cells as therapy for central nervous system disease. Exp Mol Pathol. 2015;98(2):145–51. https://doi.org/10.1016/j.yexmp.2015.01.016.

    Article  CAS  PubMed  Google Scholar 

  4. Banks WA. Drug delivery to the brain in Alzheimer’s disease: consideration of the blood-brain barrier. Adv Drug Deliv Rev. 2012;64(7):629–39. https://doi.org/10.1016/j.addr.2011.12.005.

    Article  CAS  PubMed  Google Scholar 

  5. Frey William HI. Neurologic agents for nasal administration to the brain, Ramsey Foundation; 1990.

  6. Hashizume R, Ozawa T, Gryaznov SM, Bollen AW, Lamborn KR, Frey WH 2nd, et al. New therapeutic approach for brain tumors: intranasal delivery of telomerase inhibitor GRN163. Neuro-Oncology. 2008;10(2):112–20. https://doi.org/10.1215/15228517-2007-052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Han IK, Kim MY, Byun HM, Hwang TS, Kim JM, Hwang KW, et al. Enhanced brain targeting efficiency of intranasally administered plasmid DNA: an alternative route for brain gene therapy. J Mol Med (Berl). 2007;85(1):75–83.

    Article  CAS  Google Scholar 

  8. Ma YP, Ma MM, Ge S, Guo RB, Zhang HJ, Frey WH 2nd, et al. Intranasally delivered TGF-beta1 enters brain and regulates gene expressions of its receptors in rats. Brain Res Bull. 2007;74(4):271–7. https://doi.org/10.1016/j.brainresbull.2007.06.021.

    Article  CAS  PubMed  Google Scholar 

  9. Alcala-Barraza SR, Lee MS, Hanson LR, McDonald AA, Frey WH 2nd, McLoon LK. Intranasal delivery of neurotrophic factors BDNF, CNTF, EPO, and NT-4 to the CNS. J Drug Target. 2010;18(3):179–90. https://doi.org/10.3109/10611860903318134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Broberg EK, Peltoniemi J, Nygardas M, Vahlberg T, Roytta M, Hukkanen V. Spread and replication of and immune response to gamma134.5-negative herpes simplex virus type 1 vectors in BALB/c mice. J Virol. 2004;78(23):13139–52. https://doi.org/10.1128/JVI.78.23.13139-13152.2004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bondier JR, Michel G, Propper A, Badot PM. Harmful effects of cadmium on olfactory system in mice. Inhal Toxicol. 2008;20(13):1169–77. https://doi.org/10.1080/08958370802207292.

    Article  CAS  PubMed  Google Scholar 

  12. Danielyan L, Schafer R, von Ameln-Mayerhofer A, Buadze M, Geisler J, Klopfer T, et al. Intranasal delivery of cells to the brain. Eur J Cell Biol. 2009;88(6):315–24. https://doi.org/10.1016/j.ejcb.2009.02.001.

    Article  CAS  PubMed  Google Scholar 

  13. Jiang Y, Zhu J, Xu G, Liu X. Intranasal delivery of stem cells to the brain. Expert Opin Drug Deliv. 2011;8(5):623–32. https://doi.org/10.1517/17425247.2011.566267.

    Article  CAS  PubMed  Google Scholar 

  14. Galeano C, Qiu Z, Mishra A, Farnsworth SL, Hemmi JJ, Moreira A, Edenhoffer P, Hornsby PJ. The route by which intranasally delivered stem cells enter the central nervous system. Cell Transplant. 2018;27(3):501–14. https://doi.org/10.1177/0963689718754561.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Li G, Bonamici N, Dey M, Lesniak MS, Balyasnikova IV. Intranasal delivery of stem cell-based therapies for the treatment of brain malignancies. Expert Opin Drug Deliv. 2018;15(2):163–72. https://doi.org/10.1080/17425247.2018.1378642.

    Article  CAS  PubMed  Google Scholar 

  16. Wei N, Yu SP, Gu X, Taylor TM, Song D, Liu XF, Wei L. Delayed intranasal delivery of hypoxic-preconditioned bone marrow mesenchymal stem cells enhanced cell homing and therapeutic benefits after ischemic stroke in mice. Cell Transplant. 2013;22(6):977–91. https://doi.org/10.3727/096368912X657251.

    Article  PubMed  Google Scholar 

  17. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Nasal administration of stem cells: a promising novel route to treat neonatal ischemic brain damage. Pediatr Res. 2010;68(5):419–22. https://doi.org/10.1203/PDR.0b013e3181f1c289.

    Article  PubMed  Google Scholar 

  18. Balyasnikova IV, Prasol MS, Ferguson SD, Han Y, Ahmed AU, Gutova M, Tobias AL, Mustafi D, Rincón E, Zhang L, Aboody KS, Lesniak MS. Intranasal delivery of mesenchymal stem cells significantly extends survival of irradiated mice with experimental brain tumors. Mol Ther. 2014;22(1):140–8. https://doi.org/10.1038/mt.2013.199.

    Article  CAS  PubMed  Google Scholar 

  19. Spencer D, Yu D, Morshed RA, Li G, Pituch KC, Gao DX, Bertolino N, Procissi D, Lesniak MS, Balyasnikova IV. Pharmacologic modulation of nasal epithelium augments neural stem cell targeting of glioblastoma. Theranostics. 2019;9(7):2071–83. https://doi.org/10.7150/thno.29581.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cusulin C, Monni E, Ahlenius H, Wood J, Brune JC, Lindvall O, Kokaia Z. Embryonic stem cell-derived neural stem cells fuse with microglia and mature neurons. Stem Cells. 2012;30(12):2657–71. https://doi.org/10.1002/stem.1227.

    Article  CAS  PubMed  Google Scholar 

  21. Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev. 2012;64(7):614–28. https://doi.org/10.1016/j.addr.2011.11.002.

    Article  CAS  PubMed  Google Scholar 

  22. Fortuna A, Alves G, Serralheiro A, Sousa J, Falcao A. Intranasal delivery of systemic-acting drugs: small-molecules and biomacromolecules. Eur J Pharm Biopharm. 2014;88(1):8–27. https://doi.org/10.1016/j.ejpb.2014.03.004.

    Article  CAS  PubMed  Google Scholar 

  23. Corrigan M, Wilson SS, Hampton J. Safety and efficacy of intranasally administered medications in the emergency department and prehospital settings. Am J Health Syst Pharm. 2015;72(18):1544–54. https://doi.org/10.2146/ajhp140630.

    Article  CAS  PubMed  Google Scholar 

  24. Crowe TP, Greenlee MHW, Kanthasamy AG, Hsu WH. Mechanism of intranasal drug delivery directly to the brain. Life Sci. 2018;195:44–52. https://doi.org/10.1016/j.lfs.2017.12.025.

    Article  CAS  Google Scholar 

  25. Erdo F, Bors LA, Farkas D, Bajza A, Gizurarson S. Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res Bull. 2018;143:155–70. https://doi.org/10.1016/j.brainresbull.2018.10.009.

    Article  CAS  PubMed  Google Scholar 

  26. Thorne RG, Emory CR, Ala TA, Frey WH 2nd. Quantitative analysis of the olfactory pathway for drug delivery to the brain. Brain Res. 1995;692(1–2):278–82. https://doi.org/10.1016/0006-8993(95)00637-6.

    Article  CAS  PubMed  Google Scholar 

  27. Doty RL. The olfactory vector hypothesis of neurodegenerative disease: is it viable? Ann Neurol. 2008;63(1):7–15. https://doi.org/10.1002/ana.21327.

    Article  PubMed  Google Scholar 

  28. Robertson B, Arvidsson J. Transganglionic transport of wheat germ agglutinin-HRP and choleragenoid-HRP in rat trigeminal primary sensory neurons. Brain Res. 1985;348(1):44–51. https://doi.org/10.1016/0006-8993(85)90357-9.

    Article  CAS  PubMed  Google Scholar 

  29. Anton F, Peppel P. Central projections of trigeminal primary afferents innervating the nasal mucosa: a horseradish peroxidase study in the rat. Neuroscience. 1991;41(2–3):617–28. https://doi.org/10.1016/0306-4522(91)90354-Q.

    Article  CAS  PubMed  Google Scholar 

  30. Shen L, Weber CR, Raleigh DR, Yu D, Turner JR. Tight junction pore and leak pathways: a dynamic duo. Annu Rev Physiol. 2011;73(1):283–309. https://doi.org/10.1146/annurev-physiol-012110-142150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lochhead JJ, Davis TP. Perivascular and perineural pathways involved in brain delivery and distribution of drugs after intranasal administration. Pharmaceutics. 2019;11(11):598. https://doi.org/10.3390/pharmaceutics11110598.

  32. Monje M. Myelin plasticity and nervous system function. Annu Rev Neurosci. 2018;41(1):61–76. https://doi.org/10.1146/annurev-neuro-080317-061853.

    Article  CAS  PubMed  Google Scholar 

  33. Steinke A, Meier-Stiegen S, Drenckhahn D, Asan E. Molecular composition of tight and adherens junctions in the rat olfactory epithelium and fila. Histochem Cell Biol. 2008;130(2):339–61. https://doi.org/10.1007/s00418-008-0441-8.

    Article  CAS  PubMed  Google Scholar 

  34. Thorne RG, Pronk GJ, Padmanabhan V, Frey WH 2nd. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience. 2004;127(2):481–96. https://doi.org/10.1016/j.neuroscience.2004.05.029.

    Article  CAS  PubMed  Google Scholar 

  35. Lochhead JJ, Wolak DJ, Pizzo ME, Thorne RG. Rapid transport within cerebral perivascular spaces underlies widespread tracer distribution in the brain after intranasal administration. J Cereb Blood Flow Metab. 2015;35(3):371–81. https://doi.org/10.1038/jcbfm.2014.215.

    Article  CAS  PubMed  Google Scholar 

  36. Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, Deane R, Nedergaard M. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci. 2013;33(46):18190–9. https://doi.org/10.1523/JNEUROSCI.1592-13.2013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra11.

    Article  Google Scholar 

  38. Pizzo ME, Wolak DJ, Kumar NN, Brunette E, Brunnquell CL, Hannocks MJ, Abbott NJ, Meyerand ME, Sorokin L, Stanimirovic DB, Thorne RG. Intrathecal antibody distribution in the rat brain: surface diffusion, perivascular transport and osmotic enhancement of delivery. J Physiol. 2018;596(3):445–75. https://doi.org/10.1113/JP275105.

    Article  CAS  PubMed  Google Scholar 

  39. Bovetti S, Hsieh YC, Bovolin P, Perroteau I, Kazunori T, Puche AC. Blood vessels form a scaffold for neuroblast migration in the adult olfactory bulb. J Neurosci. 2007;27(22):5976–80. https://doi.org/10.1523/JNEUROSCI.0678-07.2007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Danielyan L, Schwab M, Siegel G, Brawek B, Garaschuk O, Asavapanumas N, Buadze M, Lourhmati A, Wendel HP, Avci-Adali M, Krueger MA, Calaminus C, Naumann U, Winter S, Schaeffeler E, Spogis A, Beer-Hammer S, Neher JJ, Spohn G, Kretschmer A, Krämer-Albers EM, Barth K, Lee HJ, Kim SU, Frey WH II, Claussen CD, Hermann DM, Doeppner TR, Seifried E, Gleiter CH, Northoff H, Schäfer R. Cell motility and migration as determinants of stem cell efficacy. EBioMedicine. 2020;60:102989. https://doi.org/10.1016/j.ebiom.2020.102989.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Conti L, Cattaneo E. Neural stem cell systems: physiological players or in vitro entities? Nat Rev Neurosci. 2010;11(3):176–87. https://doi.org/10.1038/nrn2761.

    Article  CAS  PubMed  Google Scholar 

  42. Grochowski C, Radzikowska E, Maciejewski R. Neural stem cell therapy-brief review. Clin Neurol Neurosurg. 2018;173:8–14. https://doi.org/10.1016/j.clineuro.2018.07.013.

    Article  PubMed  Google Scholar 

  43. Dey M, Yu D, Kanojia D, Li G, Sukhanova M, Spencer DA, Pituch KC, Zhang L, Han Y, Ahmed AU, Aboody KS, Lesniak MS, Balyasnikova IV. Intranasal oncolytic virotherapy with CXCR4-enhanced stem cells extends survival in mouse model of glioma. Stem Cell Reports. 2016;7(3):471–82. https://doi.org/10.1016/j.stemcr.2016.07.024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Reitz M, Demestre M, Sedlacik J, Meissner H, Fiehler J, Kim SU, Westphal M, Schmidt NO. Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma. Stem Cells Transl Med. 2012;1(12):866–73. https://doi.org/10.5966/sctm.2012-0045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Dominguez-Garcia S, Geribaldi-Doldan N, Gomez-Oliva R, Ruiz FA, Carrascal L, Bolivar J, et al. A novel PKC activating molecule promotes neuroblast differentiation and delivery of newborn neurons in brain injuries. Cell Death Dis. 2020;11(4):262. https://doi.org/10.1038/s41419-020-2453-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li YH, Yu JW, Xi JY, Yu WB, Liu JC, Wang Q, Song LJ, Feng L, Yan YP, Zhang GX, Xiao BG, Ma CG. Fasudil enhances therapeutic efficacy of neural stem cells in the mouse model of MPTP-induced Parkinson’s disease. Mol Neurobiol. 2017;54(7):5400–13. https://doi.org/10.1007/s12035-016-0027-8.

    Article  CAS  PubMed  Google Scholar 

  47. Xie L, Zeng X, Hu J, Chen Q. Characterization of nestin, a selective marker for bone marrow derived mesenchymal stem cells. Stem Cells Int. 2015;2015:762098.

    Article  Google Scholar 

  48. Vaquero J, Zurita M. Functional recovery after severe CNS trauma: current perspectives for cell therapy with bone marrow stromal cells. Prog Neurobiol. 2011;93(3):341–9. https://doi.org/10.1016/j.pneurobio.2010.12.002.

    Article  PubMed  Google Scholar 

  49. Simon C, Gan QF, Kathivaloo P, Mohamad NA, Dhamodharan J, Krishnan A, et al. Deciduous DPSCs ameliorate MPTP-mediated neurotoxicity, sensorimotor coordination and olfactory function in parkinsonian mice. Int J Mol Sci. 2019;20(3):568. https://doi.org/10.3390/ijms20030568.

  50. Bagheri-Mohammadi S, Alani B, Karimian M, Moradian-Tehrani R, Noureddini M. Intranasal administration of endometrial mesenchymal stem cells as a suitable approach for Parkinson’s disease therapy. Mol Biol Rep. 2019;46(4):4293–302. https://doi.org/10.1007/s11033-019-04883-8.

    Article  CAS  PubMed  Google Scholar 

  51. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76. https://doi.org/10.1016/j.cell.2006.07.024.

    Article  CAS  PubMed  Google Scholar 

  52. Ogulur I, Gurhan G, Kombak FE, Filinte D, Barlan I, Akkoc T. Allogeneic pluripotent stem cells suppress airway inflammation in murine model of acute asthma. Int Immunopharmacol. 2014;22(1):31–40. https://doi.org/10.1016/j.intimp.2014.06.011.

    Article  CAS  PubMed  Google Scholar 

  53. Royce SG, Rele S, Broughton BRS, Kelly K, Samuel CS. Intranasal administration of mesenchymoangioblast-derived mesenchymal stem cells abrogates airway fibrosis and airway hyperresponsiveness associated with chronic allergic airways disease. FASEB J. 2017;31(9):4168–78. https://doi.org/10.1096/fj.201700178R.

    Article  CAS  PubMed  Google Scholar 

  54. Royce SG, Mao W, Lim R, Kelly K, Samuel CS. iPSC- and mesenchymoangioblast-derived mesenchymal stem cells provide greater protection against experimental chronic allergic airways disease compared with a clinically used corticosteroid. FASEB J. 2019;33(5):6402–11. https://doi.org/10.1096/fj.201802307R.

    Article  CAS  PubMed  Google Scholar 

  55. Rhea EM, Logsdon AF, Banks WA, Erickson ME. Intranasal delivery: effects on the neuroimmune axes and treatment of neuroinflammation. Pharmaceutics. 2020;12(11):1120. https://doi.org/10.3390/pharmaceutics12111120.

  56. Danielyan L, Schafer R, von Ameln-Mayerhofer A, Bernhard F, Verleysdonk S, Buadze M, et al. Therapeutic efficacy of intranasally delivered mesenchymal stem cells in a rat model of Parkinson disease. Rejuvenation Res. 2011;14(1):3–16. https://doi.org/10.1089/rej.2010.1130.

    Article  CAS  PubMed  Google Scholar 

  57. McDonald CA, Djuliannisaa Z, Petraki M, Paton MCB, Penny TR, Sutherland AE, et al. Intranasal delivery of mesenchymal stromal cells protects against neonatal hypoxic(−)ischemic brain injury. Int J Mol Sci. 2019;20(10):2449. https://doi.org/10.3390/ijms20102449.

  58. Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders--time for clinical translation? J Clin Invest. 2010;120(1):29–40. https://doi.org/10.1172/JCI40543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hayes MW, Fung VS, Kimber TE, O'Sullivan JD. Updates and advances in the treatment of Parkinson disease. Med J Aust. 2019;211(6):277–83. https://doi.org/10.5694/mja2.50224.

    Article  PubMed  Google Scholar 

  60. Jamebozorgi K, Taghizadeh E, Rostami D, Pormasoumi H, Barreto GE, Hayat SMG, Sahebkar A. Cellular and molecular aspects of Parkinson treatment: future therapeutic perspectives. Mol Neurobiol. 2019;56(7):4799–811. https://doi.org/10.1007/s12035-018-1419-8.

    Article  CAS  PubMed  Google Scholar 

  61. Bossolasco P, Cova L, Levandis G, Diana V, Cerri S, Lambertenghi Deliliers G, Polli E, Silani V, Blandini F, Armentero MT. Noninvasive near-infrared live imaging of human adult mesenchymal stem cells transplanted in a rodent model of Parkinson's disease. Int J Nanomedicine. 2012;7:435–47. https://doi.org/10.2147/IJN.S27537.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Danielyan L, Beer-Hammer S, Stolzing A, Schafer R, Siegel G, Fabian C, et al. Intranasal delivery of bone marrow-derived mesenchymal stem cells, macrophages, and microglia to the brain in mouse models of Alzheimer’s and Parkinson’s disease. Cell Transplant. 2014;23(Suppl 1):S123–39.

    Article  Google Scholar 

  63. Salama M, Sobh M, Emam M, Abdalla A, Sabry D, El-Gamal M, et al. Effect of intranasal stem cell administration on the nigrostriatal system in a mouse model of Parkinson’s disease. Exp Ther Med. 2017;13(3):976–82. https://doi.org/10.3892/etm.2017.4073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Duncan T, Valenzuela M. Alzheimer’s disease, dementia, and stem cell therapy. Stem Cell Res Ther. 2017;8(1):111. https://doi.org/10.1186/s13287-017-0567-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mita T, Furukawa-Hibi Y, Takeuchi H, Hattori H, Yamada K, Hibi H, Ueda M, Yamamoto A. Conditioned medium from the stem cells of human dental pulp improves cognitive function in a mouse model of Alzheimer’s disease. Behav Brain Res. 2015;293:189–97. https://doi.org/10.1016/j.bbr.2015.07.043.

    Article  CAS  PubMed  Google Scholar 

  66. Harach T, Jammes F, Muller C, Duthilleul N, Cheatham V, Zufferey V, Cheatham D, Lukasheva YA, Lasser T, Bolmont T. Administrations of human adult ischemia-tolerant mesenchymal stem cells and factors reduce amyloid beta pathology in a mouse model of Alzheimer’s disease. Neurobiol Aging. 2017;51:83–96. https://doi.org/10.1016/j.neurobiolaging.2016.11.009.

    Article  CAS  PubMed  Google Scholar 

  67. Perets N, Betzer O, Shapira R, Brenstein S, Angel A, Sadan T, Ashery U, Popovtzer R, Offen D. Golden exosomes selectively target brain pathologies in neurodegenerative and neurodevelopmental disorders. Nano Lett. 2019;19(6):3422–31. https://doi.org/10.1021/acs.nanolett.8b04148.

    Article  CAS  PubMed  Google Scholar 

  68. Yu-Taeger L, Stricker-Shaver J, Arnold K, Bambynek-Dziuk P, Novati A, Singer E, et al. Intranasal administration of mesenchymal stem cells ameliorates the abnormal dopamine transmission system and inflammatory reaction in the R6/2 mouse model of Huntington disease. Cells. 2019;8(6):595. https://doi.org/10.3390/cells8060595.

  69. Psaltis PJ, Simari RD. Vascular wall progenitor cells in health and disease. Circ Res. 2015;116(8):1392–412. https://doi.org/10.1161/CIRCRESAHA.116.305368.

    Article  CAS  PubMed  Google Scholar 

  70. Zhao Q, Hu J, Xiang J, Gu Y, Jin P, Hua F, et al. Intranasal administration of human umbilical cord mesenchymal stem cells-conditioned medium enhances vascular remodeling after stroke. Brain Res. 2015;1624:489–96.

    Article  CAS  Google Scholar 

  71. Nijboer CH, Kooijman E, van Velthoven CT, van Tilborg E, Tiebosch IA, Eijkelkamp N, Dijkhuizen RM, Kesecioglu J, Heijnen CJ. Intranasal stem cell treatment as a novel therapy for subarachnoid hemorrhage. Stem Cells Dev. 2018;27(5):313–25. https://doi.org/10.1089/scd.2017.0148.

    Article  CAS  PubMed  Google Scholar 

  72. Donega V, van Velthoven CT, Nijboer CH, van Bel F, Kas MJ, Kavelaars A, et al. Intranasal mesenchymal stem cell treatment for neonatal brain damage: long-term cognitive and sensorimotor improvement. PLoS One. 2013;8(1):e51253. https://doi.org/10.1371/journal.pone.0051253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Oppliger B, Joerger-Messerli M, Mueller M, Reinhart U, Schneider P, Surbek DV, Schoeberlein A. Intranasal delivery of umbilical cord-derived mesenchymal stem cells preserves myelination in perinatal brain damage. Stem Cells Dev. 2016;25(16):1234–42. https://doi.org/10.1089/scd.2016.0027.

    Article  CAS  PubMed  Google Scholar 

  74. Herz J, Koster C, Reinboth BS, Dzietko M, Hansen W, Sabir H, et al. Interaction between hypothermia and delayed mesenchymal stem cell therapy in neonatal hypoxic-ischemic brain injury. Brain Behav Immun. 2018;70:118–30. https://doi.org/10.1016/j.bbi.2018.02.006.

    Article  CAS  PubMed  Google Scholar 

  75. Wang F, Wang Z, Wang F, Dong K, Zhang J, Sun YJ, Liu CF, Xing MJ, Cheng X, Wei S, Zheng JW, Zhao XF, Wang XM, Fu J, Song HF. Comparative strategies for stem cell biodistribution in a preclinical study. Acta Pharmacol Sin. 2020;41(4):572–80. https://doi.org/10.1038/s41401-019-0313-x.

    Article  CAS  PubMed  Google Scholar 

  76. Veronesi MC, Alhamami M, Miedema SB, Yun Y, Ruiz-Cardozo M, Vannier MW. Imaging of intranasal drug delivery to the brain. Am J Nucl Med Mol Imaging. 2020;10(1):1–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Krishnan JKS, Arun P, Chembukave B, Appu AP, Vijayakumar N, Moffett JR, Puthillathu N, Namboodiri AMA. Effect of administration method, animal weight and age on the intranasal delivery of drugs to the brain. J Neurosci Methods. 2017;286:16–21. https://doi.org/10.1016/j.jneumeth.2017.05.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was funded by the Jiangsu Provincial Key R&D Program (BE2018658), Jiangsu Provincial Medical Key Discipline Project (ZDXKB2016022), Suzhou Clinical Research Center of Neurological Disease (Szzx201503), Discipline Construction Program of the Second Affiliated Hospital Soochow University (XKTJ-XK202001), and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

Author information

Authors and Affiliations

Authors

Contributions

YT Z and KJ H found the references and drafted the manuscript. KJ H and JB Z read the literature and summarized the information. QH M helped to draft the manuscript. FW designed the literature retrieval strategy. CF L modified the manuscript. FW and CF L guided the above work. The authors read and approved the final manuscript.

Corresponding authors

Correspondence to Fen Wang or Chun-Feng Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, YT., He, KJ., Zhang, JB. et al. Advances in intranasal application of stem cells in the treatment of central nervous system diseases. Stem Cell Res Ther 12, 210 (2021). https://doi.org/10.1186/s13287-021-02274-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-021-02274-0

Keywords