ORIGINAL ARTICLE

Folia Morphol.

Vol. 82, No. 4, pp. 841–853

DOI: 10.5603/FM.a2022.0097

Copyright © 2023 Via Medica

ISSN 0015–5659

eISSN 1644–3284

journals.viamedica.pl

Transplantation of bone marrow-derived mesenchymal stem cells ameliorated dopamine system impairment in a D-galactose-induced brain ageing in rats

G. El-Akabawy123S.O.F. El-Kersh4L.A. Rashed5S.N. Amin67A.A.K. El-Sheikh8
1Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman, United Arab Emirates
2Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
3Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
4Faculty of Medicine, Galala University, Suez, Egypt
5Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
6Department of Anatomy, Physiology, and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
7Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
8Basic Health Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia

[Received: 17 September 2022; Accepted: 3 November 2022; Early publication date: 29 November 2022]

Background: Ageing is the primary risk factor for Parkinson’s disease. Progressive motor and coordination decline that occurs with ageing has been linked to nigrostriatal dysfunction. Few studies have investigated the efficacy of mesenchymal stem cells in ameliorating the structural and functional alterations in the ageing nigrostriatal system. This study is the first to evaluate the effects of intravenous injection of bone marrow-derived mesenchymal stem cells (BMMSCs) in a D-galactose-induced rat model of nigrostriatal ageing.
Materials and methods: BMMSCs were intravenously injected once every 2 weeks for 8 weeks. The transplanted cells survived, migrated to the brain, and differentiated into dopaminergic neurones and astrocytes.
Results: BMMSC transplantation improved locomotor activity, restored dopaminergic system function, preserved atrophic dopaminergic neurones in the substantia nigra, exerted antioxidative effects, and restored neurotrophic factors.
Conclusions: Our findings demonstrate the efficacy of BMMSC injection in a nigrostriatal ageing rat model, and suggest that these cells may provide an effective therapeutic approach for the ageing nigrostriatal system. (Folia Morphol 2023; 82, 4: 841–853)
Key words: bone marrow-mesenchymal stem cells, D-galactose, rat, nigrostriatal dysfunction

Address for correspondence: G.F.A. El-Akabawy, PhD, Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman, United Arab Emirates, tel: 00971554200803, e-mail: g.elakabawy@ajman.ac.ae; Gehanakabawy@gmail.com

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially.

INTRODUCTION

Ageing is associated with several biochemical and molecular changes that eventually lead to cognitive and somatosensory impairments. This can be considered a primary risk factor for the development of neurodegenerative diseases such as Parkinson’s disease (PD) [8, 23, 54, 72]. Dopaminergic (DA) neurones are among the most vulnerable cells of the central nervous system to the deleterious consequences of ageing. The most obvious indication of DA neurone susceptibility to ageing is the degradation of nigrostriatal DA neurones [35, 56].

Ageing has also been associated with structural alterations of the nigrostriatal system. Several reports have shown that pathogenic alterations associated with PD are identical to age-related changes in DA neurones. The loss of DA neurones in the substantia nigra (SN), which reduces the amount of dopamine released and the number of DA receptors in the striatum and causes bradykinesia, muscular stiffness, and shaking, is a notable neuropathological aspect of PD [21, 35, 44, 56, 63]. These characteristics are mostly related to ageing (also known as late-onset PD) and are dependent on environmental and hereditary variables [19]. A substantial body of evidence suggests that brain-derived neurotrophic factor (BDNF) is essential for the survival of SN DA neurones. BDNF helps SN neurones survive in vitro and is protective against a variety of neurotoxic injuries both in vitro and in vivo [6, 40, 46]. Another neurotrophic factor, glial cell line-derived neurotrophic factor (GDNF), supports the survival of DA cells in the midbrain, increases the function of the remaining DA neurones in the SN, and inhibits degeneration and DA neuronal death [38, 41, 42]. In addition, oxidative stress appears to be a crucial risk factor for ageing-mediated neuronal and neurotransmitter changes [25, 62].

The morphological and functional alterations associated with ageing are exacerbated in age-related diseases; therefore, therapies that attenuate primary and/or secondary ageing are the main focus of ageing research [10, 32, 33]. Both preclinical and clinical trials of stem cell treatment have demonstrated its effectiveness in the treatment of Alzheimer’s disease (AD) and PD. Mesenchymal stem cells (MSCs) are the most promising type of stem cells owing to their ability to differentiate into the neuronal phenotype, secrete neurotrophic cytokines, and promote endogenous brain repair. In addition, they have immunomodulatory, neuroprotective, and angiogenic capabilities [5, 9, 26]. In rodent PD models, bone marrow-derived MSC (BMMSC) transplantation has been demonstrated to improve behavioural performance, ameliorate DA system degeneration in the SN and striatum, attenuate histopathological alterations, reduce the inflammatory response, and induce the release of neurotrophic factors [2, 20, 37, 39, 64, 67, 69]. Based on a study in 2021, a single-centre, open-label phase 1 clinical study was conducted to assess the safety and feasibility of intravenous injection of allogeneic BMMSCs delivered in escalating doses to patients with idiopathic PD [55].

Most of the research on the effectiveness of stem cells has been conducted on preclinical animal models or on patients with AD and PD, whose structural and functional brain capabilities have significantly deteriorated. It is possible that early intervention to address neuropathological changes during primary ageing will stop or at least delay the pathological processes leading to secondary ageing, thus lowering the prevalence of age-related disorders [3, 10, 32, 33, 48, 59]. Few studies have evaluated the efficacy of MSC transplantation in animal models of ageing [13, 15, 18, 29, 47, 57, 68]. Therefore, we aimed to evaluate, for the first time, the potential beneficial effects of systemic transplantation of BMMSCs on the nigrostriatal system in a D-galactose (D-gal)-induced rat model of brain ageing to evaluate their potential as a protective approach for age-related neurodegeneration.

MATERIALS AND METHODS

Animal

Thirty male Sprague Dawley rats (8 weeks old, 180–200 g) were obtained from the Theodor Bilharz Research Institute, Imbaba, Egypt, and housed in the animal facility of the Faculty of Medicine, Menoufia University, Egypt. The rats were housed in standard polycarbonate cages with 2 rats in each cage under standard laboratory settings (22 ± 5°C, 60 ± 5% humidity, and a 12-h/12-h light/dark cycle). Standard laboratory chow and tap water were provided ad libitum. All experimental procedures involving animals were approved by the Institutional Review Board of Ajman University, UAE (IRB# M-F-A-14-Mar), and the Institutional Review Board of Menoufia University, Faculty of Medicine, Egypt (IRB# 191219ANAT), and were conducted in accordance with the guidelines on the ethical use of animals in the European Community Council Directive 2010/63/EU.

BMMSC isolation and culture

Bone marrow-derived MSC were obtained from 6–8-week-old male Sprague Dawley rats, as previously reported [49]. Briefly, bone marrow plugs were harvested from the femurs and tibias of rats using a 23-gauge needle and centrifuged for 5 min at room temperature at 1800 rpm. The cell pellets were then resuspended in Dulbecco’s modified Eagle’s medium (Gibco, Carlsbad, CA, USA) containing 10% fetal bovine serum (FBS) (Gibco) and 1% penicillin-streptomycin (Gibco) and seeded at a density of 1 × 106 cells/cm in 25 cm2 cell culture flasks. Cells were incubated at 37°C in a humidified atmosphere containing 5% CO2. The culture medium was changed every 3–4 d to eliminate non-adherent haematopoietic cells. When the cells reached 70% confluence, they were harvested for 2–5 min using 0.25% Trypsin–EDTA (Sigma-Aldrich, St. Louis, MO, USA), neutralised with a complete medium, and centrifuged at 500 × g for 5 min. Cell pellets were resuspended in a complete medium. Cell viability was examined by adding equal volumes of the cell suspension and 0.4% trypan blue (Gibco) and loading 10 μL of the stained suspension into each chamber of a haemocytometer. Viable and dead cells were counted within 5 min of the sample preparation. Cells with greater than 90% viability were subcultured at a 1:3 ratio (passage 1). The cells were used at passage 4.

Flow cytometry

Cells were resuspended in staining buffer (2% FBS/phosphate buffer solution [PBS]) and surface-stained with fluorescein isothiocyanate-conjugated mouse anti-rat CD44 (BioLegend, UK), FITCH-conjugated mouse anti-rat CD90 (BD Pharmingen, USA), or PE-conjugated rabbit anti-rat CD34 (Abcam, UK) at 4°C for 30 min. Isotype-matched antibodies served as controls. Cells were analysed using an EPICS XL flow cytometer (Beckman Coulter).

Experimental design

The rats were randomly divided into three groups: control, D-gal-treated, and D-gal + BMMSC-treated (n = 10 in each group). The sample size was calculated using G Power software. D-gal (300 mg/kg; Sigma-Aldrich, St. Louis, MO, USA) was administered subcutaneously to rats in the D-gal and D-gal + BMMSCs treatment groups daily for 8 weeks. Once every 2 weeks, 1 × 106 BMMSCs labelled with the membrane-bound fluorescent marker PKH26 (Sigma-Aldrich) were intravenously delivered to rats in the D-gal + BMMSCs group.

Behavioural tests

All animals were acclimatised for 1 week after arrival to behavioural testing. Tests were conducted 1 week after the last transplantation. The test sessions were conducted between 2 PM and 5 PM. Two observers were present throughout each session and blinded to the experimental conditions.

Open-field test. The open-field test allows the simultaneous evaluation of exploration and locomotion. A box 1 m × 1 m × 50 cm in height was made of wood. The floor of the box was divided into equal areas. Each rat was positioned in the centre of the open-field arena, and the rearing frequency and number of crossings (with both forepaws) were recorded using a video camera installed 2.5 m above the box for 5 min. The box was then placed in a noiseless room under controlled illumination.

Beam walking test. Beam walking is a test of motor coordination [22]. The rats had to traverse a beam (100 cm long wooden beam, 4 cm wide, and 3 cm tall), which was hung between a start stage at one end and their home cage at the other end at a height of 80 cm, was suspended by two pillars. Foam padding was placed underneath the beam to protect the animals from injury during a fall. A line (20 cm) was drawn at the start of the beam. During the test, the rat was placed within this starting area facing its home cage, and a stopwatch was started upon release of the animal. The timer was halted when all four paws were fully placed on the finishing platform at the other end of the beam. The numbers of footsteps and faults were also recorded.

Measurement of body weight and the brain index

The general appearance of the rats, including behavioural activity, shininess, and hair coat colour, was observed daily. Body weights were assessed weekly. At the end of the experiment, the rats were anaesthetised by intraperitoneal injection of ketamine (90 mg/kg) and xylazine (15 mg/kg), and decapitated. Brains were immediately collected from all rats and weighed. Brain indices were calculated using the following formula: brain tissue weight (mg)/final body weight (g).

Assessment of oxidative stress and antioxidants indices

A spectrophotometer was used to assess the levels of malondialdehyde (MDA) and glutathione (GSH) in brain tissues. To assess the extent of lipid peroxidation, rat striata (100 mg) were homogenised in 1 mL of PBS (pH 7.0), and the MDA concentration was measured [65]. The homogenates were centrifuged after mixing with trichloroacetic acid (20%) at 5000 rpm for 15 min. The supernatants were treated with a 5% thiobarbituric acid solution before boiling in a water bath for 10 min. The absorbance at 532 nm was measured, and the MDA concentration was estimated using a standard curve. The results are expressed in nmol per mg of protein. Ellman’s method [16] was used to assess the GSH levels. A solution of dithiobis nitrobenzoate was added to the striatal tissue homogenate and incubated for 1 h. The absorbance was measured at 412 nm. A standard curve was used to measure the GSH concentration. The findings are expressed in mmol per mg of protein.

Quantitative reverse-transcription polymerase chain reaction

Total RNA was extracted from homogenised striata of rats in each group using RNeasy Purification Reagent (Qiagen, Valencia, CA, USA), according to the manufacturer’s protocol. RNA purity was assessed with a spectrophotometer; the wavelength absorption ratio (260/280 nm) was between 1.8 and 2.0 for all preparations. RNA was reverse-transcribed into cDNA using Superscript II (Gibco Life Technologies, Grand Island, NY, USA). Quantitative polymerase chain reactions were run and analysed using a StepOneTM instrument with software version 3.1 (Applied Biosystems, Foster City, CA, USA). The reaction mixtures contained SYBR Green Master Mix (Applied Biosystems), gene-specific primer pair (Table 1), cDNA, and nuclease-free water. The cycling conditions were as follows:10 min at 95°C, followed by 40 cycles of 15 s at 95°C, and 60 s at 60°C.

Table 1. List of primers used in reverse-transcription-quantitative polymerase chain reaction

Gene name

Gene accession

Primer sequence forward/Reverse 5’3’

TH

NM_012740.3

TCGGAAGCTGATTGCAGAGA

TTCCGCTGTGTATTCCACATG

DAT

NM_012694.2

CCAGCAATTCAGTGATGACATCA

CAGCATAGCCGCCAGTACAG

VMAT2

NM_013031

CGC AAA CTG ATC CTG TTC AT

5-AGA AGA TGC TTT CGC AGG TG

D1R

NM_012546.2

GGAGGACACCGAGGATGA

ATGAGGGACGATGAAATGG

D2R

NM_012547.1

TGGGTCAGAAGGGAAGG

GATGATAAAGATGAGGAGGGT

BDNF

NM_012842

TGTCCGAGGTGGTAGTACTTCATC

CATGCAACCGAAGTATGAAATAACC

VEGF

AF062644

GAGGAAAGGGAAAGGGTCAAAA

CACAGTGAACGCTCCAGGATT

GDNF

NM_019139.1

CCAGAGAATTCCAGAGGGAA

CTTCACAGGAACCGCTACAA

BETA ACTIN

NM_031144

ATTTGGCACCACACTTTCTACA

TCACGCACGATTTCCCTCTCAG

The ABI Prism sequence detection system software was used to analyse the data, and quantification was performed using Sequence Detection Software v1.7 (PE Biosystems, Foster City, CA). Relative target gene expression was calculated using the comparative cycle threshold method [34]. All values were normalised to β-actin mRNA levels.

Immunofluorescence analysis

For immunofluorescence staining, brains were dissected and fixed at 4°C for 24 h, then cryoprotected in 30% sucrose at 4°C. Serial sections (40 μm) were cut by a cryostat and stored at –20°C until use. The sections were incubated in 10% blocking solution (10% normal goat serum in 0.3% Triton X-100 in PBS) at room temperature (RT) for 1 h, then incubated at 4°C overnight in the primary antibodies rabbit anti-glial fibrillary acidic protein (GFAP) (1:1000, Abcam, Cat. #ab7260), or rabbit anti-tyrosine hydroxylase (TH) (1:500, Abcam, Cat. #ab112). The sections were then rinsed in PBS and a secondary antibody was applied (1:500, Alexa-488, Cat. #A-11034, Molecular Probes) at RT for 1 h. Finally, the sections were rinsed in PBS and mounted in Fluoroshield mounting medium with 4’,6-diamidino-2phenylindole (DAPI) (Abcam, Cat. #ab104139).

Quantitative histological assessments

Four non-overlapping images per section were randomly captured from the striatum and SN and analysed for each brain section for each marker. Immunofluorescence images were captured using a Leica DM5500 B/11888817/12 microscope equipped with a Leica DFC450C camera, using a Leica HI PLAN 10/0.25 objective. For each image, the region of interest was the field of view at a magnification of 10x. From at least three sections/rat, immunopositive cells were counted using ImageJ software (National Institutes of Health, Bethesda, Maryland, US) by a manual approach using the plugin/cell counter tool [53] and then averaged per field for each rat. The calculated numbers for the 10 animals/experimental group were used for comparison and statistical analyses. Concerning TH immunoreactive striatal fibres, the immunoreactivity of TH fibres in the striatum was measured by densitometry as described by Febbraro et al. [17]. Photos were converted to grayscale using Image J programme (1.51 version; National Institute of Health, Bethesda, MD, USA) and analysed for grey intensity after calibrating the Image J programme by assessing the optical density. Optical density values for the treatment groups are presented as a percentage of the control groups.

Statistical analysis

Data are expressed as the mean ± standard error of the mean. Normal distributions were evaluated using the D’Argostino and Pearson normality tests, and data were analysed using one-way or two-way analysis of variance followed by a post hoc Bonferroni test. Statistical significance was set at p < 0.05. Statistical analyses were performed using the GraphPad software.

RESULTS

Characterization of BMMSCs

Mesenchymal stem cells derived from the bone marrow of Sprague-Dawley rats were spindle-shaped, fibroblast-like cells after 10 days of culture. At passage 4, cells were evaluated by flow cytometry for the expression of CD90, CD44 (mesenchymal cell marker), and CD34 (haematopoietic lineage marker). More than 90% of the cells were CD90+ and CD44+, and less than 10% were CD34+ (Fig. 1). These results indicated that the cells were mostly non-haematopoietic MSCs.

Figure 1. Characterisation of the bone marrow-derived mesenchymal stem cells (BMMSCs) population. The cell-surface phenotype of the BMMSCs was assessed by flow cytometry using antibodies against CD90 (A), CD44 (B), and CD34 (C). In total, 92.3%, and 92.1% of the cells expressed CD90 and CD44, respectively, whereas only 9.5% expressed CD34; –ve — negative; +ve — positive; pop. — population; FITC — fluorescein isothiocyante; PE — phycoerythrin.
BMMSC transplantation improved the physical characteristics and brain indices

Rats in the D-gal group showed physical signs of general ageing, such as reduced activity and rough, dull, yellow hair coat with hair loss, whereas rats in the transplanted group exhibited signs of normal activity and smooth, glossy, brightly coloured hair coat, indicating that BMMSC treatment had beneficial effects on D-gal-induced ageing. In the current study, the body weights of rats in the control, D-gal, and transplanted groups were not significantly different (Fig. 2A). However, the brain index was significantly reduced in D-gal-treated rats compared with that in the control rats, whereas the brain index of the BMMSC-treated group was significantly improved compared with that in the aged rats (Fig. 2B), demonstrating that transplanted cells reversed D-gal-induced brain atrophy.

Figure 2. Body weight (A) and brain index (B) were evaluated in the control, aged (D-gal), and transplanted (D-gal + BMMSCs) rats; **p < 0.01 vs. control rats; #p < 0.01 vs. aged rats. Data are expressed as mean ± standard error of the means; n = 10/group; BMMSCs — bone marrow-derived mesenchymal stem cells.
BMMSCs recovered locomotion and motor coordination in D-gal ageing rats

Aged rats showed significantly decreased locomotor activity, as indicated by a significant decline in the number of line crossings compared with those in the control group. This was significantly improved by BMMSC injection, as indicated by the significant increase in the number of line crossings compared to the values in the ageing group (Fig. 3A). In addition, aged rats showed increased rearing, which was significantly reversed in the BMMSC-injected group (Fig. 3B). In aged rats, beam walking tests exhibited a significant increase in crossing time (Fig. 4A), foot fault number (Fig. 4B), and footstep number (Fig. 4C), with a marked reduction in velocity (Fig. 4D), when compared to control rats. These observations demonstrate marked changes in motor coordination during ageing. Interestingly, D-gal + BMMSC-treated rats showed significant improvement in these parameters.

Figure 3. Locomotion was evaluated in control, aged (D-gal), and transplanted (D-gal + BMMSCs) rats. Locomotion (horizontal locomotion and vertical rearing) were assessed for 5 min in an open-field test; A. Number of line crossing; B. Rearing frequences; ***p < 0.001 vs. control rats; ###p < 0.001 vs. aged rats. Data are expressed as means ± standard error of the means; n = 10/group; BMMSCs — bone marrow-derived mesenchymal stem cells.
Figure 4. A–D. Motor coordination was evaluated using a beam walking test in control, aged (D-gal), and transplanted (D-gal + BMMSCs) rats; ***p < 0.001 vs. control rats; #p < 0.05 and ##p < 0.01 vs. aged rats. Data are expressed as means ± standard error of the means; n = 10/group; BMMSCs — bone marrow-derived mesenchymal stem cells.
BMMSCs restored DA system function

The impact of age on several DA targets, including receptors, transporters, and relevant enzymes in the striatum, has been reported [4, 12, 21, 27, 28]. Gene expression of the main functional components of DA neurones was assessed in the different groups. Gene expression of TH for dopamine synthesis, vesicular monoamine transporter-2 (VMAT2) for dopamine transport into the vesicle, presynaptic dopamine transporter (DAT), and main postsynaptic receptors, D1 and D2, were downregulated in the striatum of aged rats compared to control rats (Fig. 5). These declines in DA markers’ expression were significantly prevented in the striatum of the D-gal + BMMSCs group.

Figure 5. Gene expression of dopaminergic system markers in the striatum in control, aged (D-gal), and transplanted (D-gal + BMMSCs) rats as measured by reverse-transcription-quantitative polymerase chain reaction; ***p < 0.001 vs. control rats; #p < 0.05, ##p < 0.01, and ###p < 0.001 vs. aged rats. Data are expressed as means ± standard error of the means; n = 10/group; BMMSCs — bone marrow-derived mesenchymal stem cells; TH — tyrosine hydroxylase; VMAT2 — vesicular monoamine transporter-2; DAT — dopamine transporter.
BMMSCs differentiated into TH-positive cells and astrocytes, and protected DA neurones in the D-gal ageing brain

To examine the underlying mechanisms by which BMMSCs improve motor deficits and restore functional DA system alterations, we first examined whether systemically administered BMMSCs homed to and survived in the brains of transplanted rats. PKH labelled BMMSCs were extensively found in all brain regions in the transplanted group. Studies have shown that age-related changes in DA neurones are comparable to the pathogenic changes observed in PD. A decline in the number of TH-positive cells has been reported in the SN of healthy aged subjects. TH staining demonstrated a significant decrease in TH-positive cells in the SN and TH fibre density in the striatum of aged rats compared to that in the controls (Fig. 6). Transplantation of BMMSCs significantly increased the number of TH neurones in the SN and TH fibre density in the striatum compared to that in aged rats. Interestingly, some of the PKH-labelled BMMSCs co-expressed TH in SN, indicating their differentiation into DA neurones. Furthermore, in the striatum, approximately 20% of the PKH-labelled BMMSCs co-expressed GFAP, indicating their differentiation into astrocytes (Fig. 7).

Figure 6. A–H. Number of tyrosine hydroxylase (TH) positive neurones in the substantia nigra (SN) and the density of the TH fibres in the striatum in control, aged (D-gal), and transplanted (D-gal + BMMSCs) rats. Scale bar = 500 μm; ***p < 0.001 vs. control rats; ##p < 0.01 vs. aged rats. Data are expressed as means ± standard error of the means; n = 10/group; BMMSCs — bone marrow-derived mesenchymal stem cells.
Figure 7. Survival and differentiation of transplanted bone marrow-derived mesenchymal stem cells (BMMSCs) into tyrosine hydroxylase (TH) positive neurones in the substantia nigra (A–D) and astrocytes in the striatum (E–H) in the transplanted (D-gal + BMMSCs) group. A number of PKH-labelled BMMSCs (red) (B, F) co-expressed TH (green) (C) and anti-glial fibrillary acidic protein (GFAP) (green) (G). The insets display the boxed area at a higher magnification. PKH-labelled cells (red) (B, F), TH-positive cells (green) (C), GFAP-positive cells (green) (G), 4’,6-diamidino-2phenylindole (DAPI)-stained nuclei (blue) (A, E), and merged images (D, H). Scale bar = 500 μm.
BMMSCs induced antioxidative effects and restored neurotrophic factors

The modification of neuronal and neurotransmitter functions that accompanies ageing seems to be linked to oxidative stress. The specific susceptibility of SN neurones to ageing accumulated reactive oxygen species that may be the cause of the age-associated reduction in DA and motor function in elderly rats [25, 62]. Aged rats had higher levels of MDA, an index of lipid peroxidation, in the striatum than control rats. Furthermore, GSH levels were significantly downregulated in aged rats compared with those in control rats. In D-gal + BMMSC-treated rats, MDA levels significantly declined, whereas GSH levels were upregulated when compared with the levels in aged rats (Fig. 8A, B). Neurotrophic factors, such as BDNF and GDNF, are crucial for the synaptic activity, survival, and function of DA neurones, and their decline has been linked to motor deficits associated with aged animals [7, 11, 14, 24, 38, 43, 52, 71]. In the striatum of aged rats, the expression of BDNF, GDNF, and vascular endothelial growth factor (VEGF) was remarkably reduced when compared with the expression in control rats, and these declines were reversed by BMMSC transplantation (Fig. 8C).

Figure 8. Status of malondialdehyde (MDA) (A) and glutathione (GSH) (B) and gene expression of brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and vascular endothelial growth factor (VEGF) (C) in the striatum of control, aged (D-gal), and transplanted (D-gal + BMMSCs) rats; ***p < 0.001 vs. control rats; ##p < 0.01 and ###p < 0.001 vs. aged rats. Data are expressed as means ± standard error of the means; n = 10/group; BMMSCs — bone marrow-derived mesenchymal stem cells.

DISCUSSION

The most important risk factor for PD is ageing, and progressive motor and coordination deterioration associated with normal ageing has been linked to nigrostriatal degeneration. Several studies have shown that the DA system, both structurally and functionally, is affected during normal ageing [8, 23, 35, 44, 54, 56, 63, 72]. The capability of stem cells to replace lost or malfunctioning cells has been the focus of recent research [13, 15, 18, 29, 47, 57, 68]. This study is the first to examine the potential beneficial effects of systemic transplantation of BMMSCs on the DA system in a D-gal-induced rat model of brain ageing. In this study, we demonstrated that intravenous transplantation of BMMSCs into D-gal-aged rats ameliorated behavioural deficits, restored DA dysfunction, differentiated into TH-positive cells in the SN, protected TH immunoreactivity in the aged striatum, and induced antioxidative and neurotrophic effects. These data demonstrate the therapeutic effectiveness of BMMSCS in the aged brain.

Ageing is characterised by a gradual decline in locomotion and motor coordination. In our study, BMMSCs improved motor alterations in D-gal + BMMSCs. Our results are consistent with those of previous studies. Implantation of BMMCs into the striatum of naturally aged rats increased the functional recovery of swimming performance as measured by the Marshall scale for vigour and success, as well as motor coordination as measured by the transverse bridge test [18]. In various animal models of PD, BMMSCs implantation improved motor impairments [20, 31, 37, 45, 64, 67]. The beam walk test, which examines limb movements, such as accurate stepping, coordination, and precise positioning of the paw, is particularly susceptible to DA depletion [37, 51, 61]. In our study, the detected decrease in striatal DA levels resulted in significant motor incoordination in D-gal rats, as indicated by an increased number of footsteps and foot slips and a significant decline in velocity. Intravenous injection of BMMSCs decreased the number of footsteps and foot faults with an increase in velocity. The observed motor improvements were associated with increased DA and TH levels in the striatum, suggesting that BMMC transplantation improved motor dysfunction in aged rats, possibly through DA upregulation in the striatum.

Several studies have demonstrated the significance of DA signalling in the maintenance of motor function, and that declines in dopamine functional component availability might be responsible for age-related behavioural deficits [4, 12, 21, 27, 28]. In the treated group, the gene expression of DA markers demonstrated an overall increase compared to that in aged rats. A study evaluating the striatal implantation of rat adult bone marrow MSCs in a 6-hydroxydopamine rat model of PD reported a partial albeit significant recovery of DA presynaptic markers such as D1, D2, DAT, and VMAT2 in treated animals compared to non-treated ones [12]. Therefore, it can be concluded that the detected improvements in motor and coordination activities might be due to the recovery of the DA system.

The survival, migration, and differentiation capacity of the injected BMMSCs in D-gal-aged rats were investigated to explain the mechanism behind the behavioural and DA functional improvements observed following BMMSC transplantation. Repeated intravenous BMMSC injection resulted in significant cell migration across all brain areas studied. These findings corroborate prior findings. Ageing has been associated with increased blood-brain barrier permeability in both animals and humans, which might be caused by numerous ageing-related processes, including increased oxidative stress and greater microglial activation [50, 60]. In our study, a few transplanted cells differentiated into TH+ cells in SN. Substantial evidence suggests that BMMSCs can differentiate into neurones, particularly DA neurones, both in vitro and in vivo [1, 30, 58, 66, 70]. Moreover, the nigrostriatal pathway is known to be involved in ageing and PD progression, and the striatum is well established to be the target brain structure for DA projections from the SN. We further assessed whether the DA fibres of the striatum were re-innervated in the transplanted group. Our results revealed that transplantation of BMMSCs significantly increased the number of endogenous TH neurones in the SN and TH fibre density in the striatum compared to that in aged rats. These results suggest endogenous restoration of the host DA system in the SN of transplanted rats. Therefore, we next sought to investigate the potential paracrine mechanisms mediated by transplanted BMMSCs that could contribute to the endogenous revival of the host DA system.

Mesenchymal stem cells can be considered as mini-bioreactors capable of secreting a wide range of cytokines and neurotrophic factors that are crucial in the treatment of neurodegenerative disorders. There is a substantial body of evidence that MSCs express a variety of neurotrophic factors, including nerve growth factor, GDNF, BDNF, insulin-like growth factor-1, and basic fibroblast growth factor at both the mRNA and protein levels [24, 52, 71]. Synaptic plasticity, as well as the survival and function of midbrain dopamine neurones, are dependent on BDNF. By comparing BDNF (+/–) with wild-type mice at various ages, the effects of a partial genetic deletion of BDNF on motor activities and DA level measurements were studied. With age, a decrease in BDNF expression becomes more important for DA circuits and associated behavioural performance [11]. Depletion of BDNF leads to declined TH expression in the SN [14]. GDNF is a secretory protein that protects DA neurones both in vitro and in vivo. GDNF treatment increases striatal dopamine levels and potentiates striatal DA fibre regeneration in preclinical animals [38]. As a result, intracranial ectopic administration of GDNF has been attempted in multiple PD clinical studies with promising but equivocal outcomes [7]. We observed decreased BDNF and GDNF expression in D-gal aged rats, and these decreases were remarkably restored in BMMSC-transplanted rats. GDNF is primarily produced by astrocytes in the brain. GDNF expression is increased in astrocytes in the striatal region of PD animal models with DA innervation, reflecting a process of endogenous regeneration [43]. Interestingly, in the present study, 20% of transplanted cells differentiated into astrocytes in the striatum. The biological features of BMMSC production of neurotrophic factors such as GDNF, as well as the ability of these cells to differentiate into astrocytes, indicate their potential for treating age-related neurodegenerative diseases.

CONCLUSIONS

This study demonstrated that intravenous transplantation of BMMSCs prevented locomotion and coordination deficits in a D-gal ageing rat model by restoring DA system function, protecting atrophic DA neurones in the SN, inducing antioxidative effects, and secreting neurotrophic factors. Our study provides proof of principle that the systemic transplantation of BMMSCs is a potential therapeutic approach for the protection of nigrostriatal changes associated with ageing.

Funding

This research project was funded by the Deanship of Research and Graduate Studies, Ajman University, UAE (grant no. GL5211642).

Conflict of interest: None declared

REFERENCES

  1. Ababneh NA, Al-Kurdi B, Jamali F, et al. A comparative study of the capability of MSCs isolated from different human tissue sources to differentiate into neuronal stem cells and dopaminergic-like cells. PeerJ. 2022; 10: e13003, doi: 10.7717/peerj.13003, indexed in Pubmed: 35341051.
  2. Abdelwahab SA, Elsebay SA, Ibrahim MF, et al. Cerebral and cerebellar histological changes in the rat animal model of rotenone induced parkinsonism can be ameliorated by bone marrow derived stem cell conditioned media. J Chem Neuroanat. 2021; 111: 101892, doi: 10.1016/j.jchemneu.2020.101892, indexed in Pubmed: 33220428.
  3. Artegiani B, Calegari F. Age-related cognitive decline: can neural stem cells help us? Aging (Albany NY). 2012; 4(3): 176–186, doi: 10.18632/aging.100446, indexed in Pubmed: 22466406.
  4. Bäckman L, Nyberg L, Lindenberger U, et al. The correlative triad among aging, dopamine, and cognition: current status and future prospects. Neurosci Biobehav Rev. 2006; 30(6): 791–807, doi: 10.1016/j.neubiorev.2006.06.005, indexed in Pubmed: 16901542.
  5. Badyra B, Sułkowski M, Milczarek O, et al. Mesenchymal stem cells as a multimodal treatment for nervous system diseases. Stem Cells Transl Med. 2020; 9(10): 1174–1189, doi: 10.1002/sctm.19-0430, indexed in Pubmed: 32573961.
  6. Baquet ZC, Bickford PC, Jones KR. Brain-derived neurotrophic factor is required for the establishment of the proper number of dopaminergic neurons in the substantia nigra pars compacta. J Neurosci. 2005; 25(26): 6251–6259, doi: 10.1523/JNEUROSCI.4601-04.2005, indexed in Pubmed: 15987955.
  7. Barker RA, Björklund A, Gash DM, et al. GDNF and Parkinson’s disease: where next? A summary from a recent workshop. J Parkinsons Dis. 2020; 10(3): 875–891, doi: 10.3233/JPD-202004, indexed in Pubmed: 32508331.
  8. Behfar Q, Ramirez Zuniga A, Martino-Adami PV. Aging, Senescence, and Dementia. J Prev Alzheimers Dis. 2022; 9(3): 523–531, doi: 10.14283/jpad.2022.42, indexed in Pubmed: 35841253.
  9. Berebichez-Fridman R, Montero-Olvera PR. Sources and clinical applications of mesenchymal stem cells: state-of-the-art review. Sultan Qaboos Univ Med J. 2018; 18(3): e264–e277, doi: 10.18295/squmj.2018.18.03.002, indexed in Pubmed: 30607265.
  10. Blinkouskaya Y, Caçoilo A, Gollamudi T, et al. Brain aging mechanisms with mechanical manifestations. Mech Ageing Dev. 2021; 200: 111575, doi: 10.1016/j.mad.2021.111575, indexed in Pubmed: 34600936.
  11. Boger HA, Mannangatti P, Samuvel DJ, et al. Effects of brain-derived neurotrophic factor on dopaminergic function and motor behavior during aging. Genes Brain Behav. 2011; 10(2): 186–198, doi: 10.1111/j.1601-183X.2010.00654.x, indexed in Pubmed: 20860702.
  12. Bouchez G, Sensebé L, Vourc’h P, et al. Partial recovery of dopaminergic pathway after graft of adult mesenchymal stem cells in a rat model of Parkinson’s disease. Neurochem Int. 2008; 52(7): 1332–1342, doi: 10.1016/j.neuint.2008.02.003, indexed in Pubmed: 18372079.
  13. Cao N, Liao T, Liu J, et al. Clinical-grade human umbilical cord-derived mesenchymal stem cells reverse cognitive aging via improving synaptic plasticity and endogenous neurogenesis. Cell Death Dis. 2017; 8(8): e2996, doi: 10.1038/cddis.2017.316, indexed in Pubmed: 28796260.
  14. Dluzen DE, McDermott JL, Anderson LI, et al. Age-related changes in nigrostriatal dopaminergic function are accentuated in +/- brain-derived neurotrophic factor mice. Neuroscience. 2004; 128(1): 201–208, doi: 10.1016/j.neuroscience.2004.06.032, indexed in Pubmed: 15450367.
  15. El-Akabawy G, Aabed K, Rashed LA, et al. Preventive effects of bone marrow-derived mesenchymal stem cell transplantation in a D-galactose-induced brain aging in rats. Folia Morphol. 2022; 81(3): 632–649, doi: 10.5603/FM.a2021.0073, indexed in Pubmed: 34355788.
  16. Ellman GL. Tissue sulfhydryl groups. Arch Biochem Biophys. 1959; 82(1): 70–77, doi: 10.1016/0003-9861(59)90090-6, indexed in Pubmed: 13650640.
  17. Febbraro F, Andersen KJ, Sanchez-Guajardo V, et al. Chronic intranasal deferoxamine ameliorates motor defects and pathology in the α-synuclein rAAV Parkinson’s model. Exp Neurol. 2013; 247: 45–58, doi: 10.1016/j.expneurol.2013.03.017, indexed in Pubmed: 23531432.
  18. Fernández CI, Alberti E, Mendoza Y, et al. Motor and cognitive recovery induced by bone marrow stem cells grafted to striatum and hippocampus of impaired aged rats: functional and therapeutic considerations. Ann NY Acad Sci. 2004; 1019: 48–52, doi: 10.1196/annals.1297.011, indexed in Pubmed: 15246993.
  19. Foltynie T, Sawcer S, Brayne C, et al. The genetic basis of Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2002; 73(4): 363–370, doi: 10.1136/jnnp.73.4.363, indexed in Pubmed: 12235301.
  20. Ghahari L, Safari M, Rahimi Jaberi K, et al. Mesenchymal stem cells with granulocyte colony-stimulating factor reduce stress oxidative factors in Parkinson’s disease. Iran Biomed J. 2020; 24(2): 89–98, doi: 10.29252/ibj.24.2.89, indexed in Pubmed: 31677610.
  21. Giacobbo BL, Özalay Ö, Mediavilla T, et al. The aged striatum: evidence of molecular and structural changes using a longitudinal multimodal approach in mice. Front Aging Neurosci. 2022; 14: 795132, doi: 10.3389/fnagi.2022.795132, indexed in Pubmed: 35140600.
  22. Goldstein LB. Rapid reliable measurement of lesion parameters for studies of motor recovery after sensorimotor cortex injury in the rat. J Neurosci Methods. 1993; 48(1-2): 35–42, doi: 10.1016/s0165-0270(05)80005-6, indexed in Pubmed: 8377521.
  23. Hou Y, Dan X, Babbar M, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019; 15(10): 565–581, doi: 10.1038/s41582-019-0244-7, indexed in Pubmed: 31501588.
  24. Inden M, Takata K, Nishimura K, et al. Therapeutic effects of human mesenchymal and hematopoietic stem cells on rotenone-treated parkinsonian mice. J Neurosci Res. 2013; 91(1): 62–72, doi: 10.1002/jnr.23128, indexed in Pubmed: 23073839.
  25. Ionescu-Tucker A, Cotman CW. Emerging roles of oxidative stress in brain aging and Alzheimer’s disease. Neurobiol Aging. 2021; 107: 86–95, doi: 10.1016/j.neurobiolaging.2021.07.014, indexed in Pubmed: 34416493.
  26. Jiménez-Acosta MA, Hernández LJ, Cristerna ML, et al. Mesenchymal stem cells: new alternatives for nervous system disorders. Curr Stem Cell Res Ther. 2023; 18(3): 299–321, doi: 10.2174/1574888X17666220511153133, indexed in Pubmed: 35546750.
  27. Juarez EJ, Castrellon JJ, Green MA, et al. Reproducibility of the correlative triad among aging, dopamine receptor availability, and cognition. Psychol Aging. 2019; 34(7): 921–932, doi: 10.1037/pag0000403, indexed in Pubmed: 31589058.
  28. Karalija N, Papenberg G, Wåhlin A, et al. Sex differences in dopamine integrity and brain structure among healthy older adults: Relationships to episodic memory. Neurobiol Aging. 2021; 105: 272–279, doi: 10.1016/j.neurobiolaging.2021.04.022, indexed in Pubmed: 34134056.
  29. Kim D, Kyung J, Park D, et al. Health span-extending activity of human amniotic membrane- and adipose tissue-derived stem cells in F344 rats. Stem Cells Transl Med. 2015; 4(10): 1144–1154, doi: 10.5966/sctm.2015-0011, indexed in Pubmed: 26315571.
  30. Li M, Yang J, Cheng O, et al. Effect of TO901317 on GF to promote the differentiation of human bone marrow mesenchymal stem cells into dopamine neurons on Parkinson’s disease. Ther Adv Chronic Dis. 2021; 12: 2040622321998139, doi: 10.1177/2040622321998139, indexed in Pubmed: 33796244.
  31. Li Y, Li Z, Gu J, et al. Exosomes isolated during dopaminergic neuron differentiation suppressed neuronal inflammation in a rodent model of Parkinson’s disease. Neurosci Lett. 2022; 771: 136414, doi: 10.1016/j.neulet.2021.136414, indexed in Pubmed: 34954117.
  32. Li Z, Zhang Z, Ren Y, et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology. 2021; 22(2): 165–187, doi: 10.1007/s10522-021-09910-5, indexed in Pubmed: 33502634.
  33. Limke TL, Rao MS. Neural stem cells in aging and disease. J Cell Mol Med. 2002; 6(4): 475–496, doi: 10.1111/j.1582-4934.2002.tb00451.x, indexed in Pubmed: 12611637.
  34. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001; 25(4): 402–408, doi: 10.1006/meth.2001.1262, indexed in Pubmed: 11846609.
  35. Lopez-Leon M, Reggiani PC, Claudia B, et al. Regenerative medicine for the aging brain. Enliven: J Stem Cell Res Regen Med. 2014; 01(01), doi: 10.18650/2379-5751.11001.
  36. Lövdén M, Karalija N, Andersson M, et al. Latent-Profile analysis reveals behavioral and brain correlates of dopamine-cognition associations. Cereb Cortex. 2018; 28(11): 3894–3907, doi: 10.1093/cercor/bhx253, indexed in Pubmed: 29028935.
  37. Mahendru D, Jain A, Bansal S, et al. Neuroprotective effect of bone marrow-derived mesenchymal stem cell secretome in 6-OHDA-induced Parkinson’s disease. Regen Med. 2021; 16(10): 915–930, doi: 10.2217/rme-2021-0018, indexed in Pubmed: 34553608.
  38. Manfredsson FP, Polinski NK, Subramanian T, et al. The future of GDNF in Parkinson’s disease. Front Aging Neurosci. 2020; 12: 593572, doi: 10.3389/fnagi.2020.593572, indexed in Pubmed: 33364933.
  39. Mendes-Pinheiro B, Anjo SI, Manadas B, et al. Bone marrow mesenchymal stem cells’ secretome exerts neuroprotective effects in a Parkinson’s disease rat model. Front Bioeng Biotechnol. 2019; 7: 294, doi: 10.3389/fbioe.2019.00294, indexed in Pubmed: 31737616.
  40. Mercado NM, Collier TJ, Sortwell CE, et al. BDNF in the Aged Brain: Translational Implications for Parkinson’s Disease. Austin Neurol Neurosci. 2017; 2(2), indexed in Pubmed: 29726549.
  41. Mickiewicz AL, Kordower JH. GDNF family ligands: a potential future for Parkinson’s disease therapy. CNS Neurol Disord Drug Targets. 2011; 10(6): 703–711, doi: 10.2174/187152711797247876, indexed in Pubmed: 21838676.
  42. Mitra S, Turconi G, Darreh-Shori T, et al. Increased endogenous GDNF in mice protects against age-related decline in neuronal cholinergic markers. Front Aging Neurosci. 2021; 13: 714186, doi: 10.3389/fnagi.2021.714186, indexed in Pubmed: 34475820.
  43. Nakagawa T, Yabe T, Schwartz JP. Gene expression profiles of reactive astrocytes cultured from dopamine-depleted striatum. Neurobiol Dis. 2005; 20(2): 275–282, doi: 10.1016/j.nbd.2005.03.009, indexed in Pubmed: 16242635.
  44. Naoi M, Maruyama W. Cell death of dopamine neurons in aging and Parkinson’s disease. Mech Ageing Dev. 1999; 111(2-3): 175–188, doi: 10.1016/s0047-6374(99)00064-0, indexed in Pubmed: 10656535.
  45. Nicaise AM, Willis CM, Crocker SJ, et al. Stem cells of the aging brain. Front Aging Neurosci. 2020; 12: 247, doi: 10.3389/fnagi.2020.00247, indexed in Pubmed: 32848716.
  46. Palasz E, Wysocka A, Gasiorowska A, et al. BDNF as a promising therapeutic agent in parkinson’s disease. Int J Mol Sci. 2020; 21(3), doi: 10.3390/ijms21031170, indexed in Pubmed: 32050617.
  47. Park D, Yang G, Bae DK, et al. Human adipose tissue-derived mesenchymal stem cells improve cognitive function and physical activity in ageing mice. J Neurosci Res. 2013; 91(5): 660–670, doi: 10.1002/jnr.23182, indexed in Pubmed: 23404260.
  48. Piccardi L, Curcio G, Palermo L, et al. Ageing and neurodegenerative disorders. Behav Neurol. 2015; 2015: 149532, doi: 10.1155/2015/149532, indexed in Pubmed: 26185358.
  49. Polisetti N, Chaitanya VG, Babu PP, et al. Isolation, characterization and differentiation potential of rat bone marrow stromal cells. Neurol India. 2010; 58(2): 201–208, doi: 10.4103/0028-3886.63789, indexed in Pubmed: 20508336.
  50. Popescu BO, Toescu EC, Popescu LM, et al. Blood-brain barrier alterations in ageing and dementia. J Neurol Sci. 2009; 283(1-2): 99–106, doi: 10.1016/j.jns.2009.02.321, indexed in Pubmed: 19264328.
  51. Quinn LP, Perren MJ, Brackenborough KT, et al. A beam-walking apparatus to assess behavioural impairments in MPTP-treated mice: pharmacological validation with R-(-)-deprenyl. J Neurosci Methods. 2007; 164(1): 43–49, doi: 10.1016/j.jneumeth.2007.03.021, indexed in Pubmed: 17498809.
  52. Rahbaran M, Zekiy AO, Bahramali M, et al. Therapeutic utility of mesenchymal stromal cell (MSC)-based approaches in chronic neurodegeneration: a glimpse into underlying mechanisms, current status, and prospects. Cell Mol Biol Lett. 2022; 27(1): 56, doi: 10.1186/s11658-022-00359-z, indexed in Pubmed: 35842587.
  53. Rangan GK, Tesch GH. Quantification of renal pathology by image analysis. Nephrology (Carlton). 2007; 12(6): 553–558, doi: 10.1111/j.1440-1797.2007.00855.x, indexed in Pubmed: 17995580.
  54. Reeve A, Simcox E, Turnbull D. Ageing and Parkinson’s disease: why is advancing age the biggest risk factor? Ageing Res Rev. 2014; 14(100): 19–30, doi: 10.1016/j.arr.2014.01.004, indexed in Pubmed: 24503004.
  55. Schiess M, Suescun J, Doursout MF, et al. Allogeneic bone marrow-derived mesenchymal stem cell safety in idiopathic Parkinson’s disease. Mov Disord. 2021; 36(8): 1825–1834, doi: 10.1002/mds.28582, indexed in Pubmed: 33772873.
  56. Seo JP, Koo DK. Aging of the nigrostriatal tract in the human brain: a diffusion tensor imaging study. Medicina (Kaunas). 2021; 57(9), doi: 10.3390/medicina57090994, indexed in Pubmed: 34577917.
  57. Shen J, Tsai YT, Dimarco NM, et al. Transplantation of mesenchymal stem cells from young donors delays aging in mice. Sci Rep. 2011; 1: 67, doi: 10.1038/srep00067, indexed in Pubmed: 22355586.
  58. Shetty P, Ravindran G, Sarang S, et al. Clinical grade mesenchymal stem cells transdifferentiated under xenofree conditions alleviates motor deficiencies in a rat model of Parkinson’s disease. Cell Biol Int. 2009; 33(8): 830–838, doi: 10.1016/j.cellbi.2009.05.002, indexed in Pubmed: 19465139.
  59. Sikora E, Bielak-Zmijewska A, Dudkowska M, et al. Cellular senescence in brain aging. Front Aging Neurosci. 2021; 13: 646924, doi: 10.3389/fnagi.2021.646924, indexed in Pubmed: 33732142.
  60. Simpson JE, Wharton SB, Cooper J, et al. Alterations of the blood-brain barrier in cerebral white matter lesions in the ageing brain. Neurosci Lett. 2010; 486(3): 246–251, doi: 10.1016/j.neulet.2010.09.063, indexed in Pubmed: 20887772.
  61. Strome EM, Cepeda IL, Sossi V, et al. Evaluation of the integrity of the dopamine system in a rodent model of Parkinson’s disease: small animal positron emission tomography compared to behavioral assessment and autoradiography. Mol Imaging Biol. 2006; 8(5): 292–299, doi: 10.1007/s11307-006-0051-6, indexed in Pubmed: 16897319.
  62. Trist BG, Hare DJ, Double KL. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell. 2019; 18(6): e13031, doi: 10.1111/acel.13031, indexed in Pubmed: 31432604.
  63. Umegaki H, Roth GS, Ingram DK. Aging of the striatum: mechanisms and interventions. Age (Dordr). 2008; 30(4): 251–261, doi: 10.1007/s11357-008-9066-z, indexed in Pubmed: 19424849.
  64. Wang X, Zhuang W, Fu W, et al. The lentiviral-mediated Nurr1 genetic engineering mesenchymal stem cells protect dopaminergic neurons in a rat model of Parkinson’s disease. Am J Transl Res. 2018; 10(6): 1583–1599, indexed in Pubmed: 30018702.
  65. Wills ED. Evaluation of lipid peroxidation in lipids and biologicalmembranes. In: Snell K, Mullock B (eds.), Biochemical Toxicology: A PracticalApproach. Oxford, London 1987.
  66. Xiong N, Yang H, Liu L, et al. bFGF promotes the differentiation and effectiveness of human bone marrow mesenchymal stem cells in a rotenone model for Parkinson’s disease. Environ Toxicol Pharmacol. 2013; 36(2): 411–422, doi: 10.1016/j.etap.2013.05.005, indexed in Pubmed: 23770451.
  67. Xue J, Liu Y, Darabi MA, et al. An injectable conductive Gelatin-PANI hydrogel system serves as a promising carrier to deliver BMSCs for Parkinson’s disease treatment. Mater Sci Eng C Mater Biol Appl. 2019; 100: 584–597, doi: 10.1016/j.msec.2019.03.024, indexed in Pubmed: 30948095.
  68. Zappa Villar MF, Lehmann M, García MG, et al. Mesenchymal stem cell therapy improves spatial memory and hippocampal structure in aging rats. Behav Brain Res. 2019; 374: 111887, doi: 10.1016/j.bbr.2019.04.001, indexed in Pubmed: 30951751.
  69. Zarbakhsh S, Safari M, Aldaghi MR, et al. Irisin protects the substantia nigra dopaminergic neurons in the rat model of Parkinson’s disease. Iran J Basic Med Sci. 2019; 22(7): 722–728, doi: 10.22038/ijbms.2019.33444.7987, indexed in Pubmed: 32373292.
  70. Zhang J, Yang Bo, Luo L, et al. Effect of NTN and Lmx1 on the notch signaling pathway during the differentiation of human bone marrow mesenchymal stem cells into dopaminergic neuron-like cells. Parkinsons Dis. 2021; 2021: 6676709, doi: 10.1155/2021/6676709, indexed in Pubmed: 34373779.
  71. Zheng W, Honmou O, Miyata K, et al. Therapeutic benefits of human mesenchymal stem cells derived from bone marrow after global cerebral ischemia. Brain Res. 2010; 1310: 8–16, doi: 10.1016/j.brainres.2009.11.012, indexed in Pubmed: 19913518.
  72. Zia A, Pourbagher-Shahri AM, Farkhondeh T, et al. Molecular and cellular pathways contributing to brain aging. Behav Brain Funct. 2021; 17(1): 6, doi: 10.1186/s12993-021-00179-9, indexed in Pubmed: 34118939.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., Grupa Via Medica, Świętokrzyska 73, 80–180 Gdańsk, Poland

tel.: +48 58 320 94 94, faks: +48 58 320 94 60, e-mail: viamedica@viamedica.pl