Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cells ; 13(6)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38534313

ABSTRACT

Huntington's disease (HD), a congenital neurodegenerative disorder, extends its pathological damages beyond the nervous system. The systematic manifestation of HD has been extensively described in numerous studies, including dysfunction in peripheral organs and peripheral inflammation. Gut dysbiosis and the gut-liver-brain axis have garnered greater emphasis in neurodegenerative research, and increased plasma levels of pro-inflammatory cytokines have been identified in HD patients and various in vivo models, correlating with disease progression. In the present study, we investigated hepatic pathological markers in the liver of R6/2 mice which convey exon 1 of the human mutant huntingtin gene. Furthermore, we evaluated the impact of intravenously administered Mesenchymal Stromal Cells (MSCs) on the liver enzymes, changes in hepatic inflammatory markers, as well as brain pathology and behavioral deficits in R6/2 mice. Our results revealed altered enzyme expression and increased levels of inflammatory mediators in the liver of R6/2 mice, which were significantly attenuated in the MSC-treated R6/2 mice. Remarkably, neuronal pathology and altered motor activities in the MSC-treated R6/2 mice were significantly ameliorated, despite the absence of MSCs in the postmortem brain. Our data highlight the importance of hepatic pathological changes in HD, providing a potential therapeutic approach. Moreover, the data open new perspectives for the search in blood biomarkers correlating with liver pathology in HD.


Subject(s)
Huntington Disease , Mice , Humans , Animals , Huntington Disease/metabolism , Mice, Transgenic , Disease Models, Animal , Brain/metabolism , Liver/metabolism
2.
Adv Sci (Weinh) ; 11(18): e2307734, 2024 May.
Article in English | MEDLINE | ID: mdl-38430535

ABSTRACT

The hepatic content of amyloid beta (Aß) decreases drastically in human and rodent cirrhosis highlighting the importance of understanding the consequences of Aß deficiency in the liver. This is especially relevant in view of recent advances in anti-Aß therapies for Alzheimer's disease (AD). Here, it is shown that partial hepatic loss of Aß in transgenic AD mice immunized with Aß antibody 3D6 and its absence in amyloid precursor protein (APP) knockout mice (APP-KO), as well as in human liver spheroids with APP knockdown upregulates classical hallmarks of fibrosis, smooth muscle alpha-actin, and collagen type I. Aß absence in APP-KO and deficiency in immunized mice lead to strong activation of transforming growth factor-ß (TGFß), alpha secretases, NOTCH pathway, inflammation, decreased permeability of liver sinusoids, and epithelial-mesenchymal transition. Inversely, increased systemic and intrahepatic levels of Aß42 in transgenic AD mice and neprilysin inhibitor LBQ657-treated wild-type mice protect the liver against carbon tetrachloride (CCl4)-induced injury. Transcriptomic analysis of CCl4-treated transgenic AD mouse livers uncovers the regulatory effects of Aß42 on mitochondrial function, lipid metabolism, and its onco-suppressive effects accompanied by reduced synthesis of extracellular matrix proteins. Combined, these data reveal Aß as an indispensable regulator of cell-cell interactions in healthy liver and a powerful protector against liver fibrosis.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Liver , Mice, Transgenic , Animals , Mice , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Liver/metabolism , Liver/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Humans , Mice, Knockout , Mice, Inbred C57BL
3.
Mol Oncol ; 18(3): 528-546, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38115217

ABSTRACT

Neural stem cells (NSCs) are considered to be valuable candidates for delivering a variety of anti-cancer agents, including oncolytic viruses, to brain tumors. However, owing to the previously reported tumorigenic potential of NSC cell lines after intranasal administration (INA), here we identified the human hepatic stellate cell line LX-2 as a cell type capable of longer resistance to replication of oncolytic adenoviruses (OAVs) as a therapeutic cargo, and that is non-tumorigenic after INA. Our data show that LX-2 cells can longer withstand the OAV XVir-N-31 replication and oncolysis than NSCs. By selecting the highly migratory cell population out of LX-2, an offspring cell line with a higher and more stable capability to migrate was generated. Additionally, as a safety backup, we applied genomic herpes simplex virus thymidine kinase (HSV-TK) integration into LX-2, leading to high vulnerability to ganciclovir (GCV). Histopathological analyses confirmed the absence of neoplasia in the respiratory tracts and brains of immuno-compromised mice 3 months after INA of LX-2 cells. Our data suggest that LX-2 is a novel, robust, and safe cell line for delivering anti-cancer and other therapeutic agents to the brain.


Subject(s)
Antiviral Agents , Genetic Therapy , Mice , Humans , Animals , Administration, Intranasal , Cell Line , Central Nervous System/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Thymidine Kinase/therapeutic use
4.
Cancers (Basel) ; 15(20)2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37894279

ABSTRACT

A glioblastoma (GBM) is an aggressive and lethal primary brain tumor with restricted treatment options and a dismal prognosis. Oncolytic virotherapy (OVT) has developed as a promising approach for GBM treatment. However, reaching invasive GBM cells may be hindered by tumor-surrounding, non-neoplastic cells when the oncolytic virus (OV) is applied intratumorally. Using two xenograft GBM mouse models and immunofluorescence analyses, we investigated the intranasal delivery of the oncolytic adenovirus (OAV) XVir-N-31 via virus-loaded, optimized shuttle cells. Intranasal administration (INA) was selected due to its non-invasive nature and the potential to bypass the blood-brain barrier (BBB). Our findings demonstrate that the INA of XVir-N-31-loaded shuttle cells successfully delivered OAVs to the core tumor and invasive GBM cells, significantly prolonged the survival of the GBM-bearing mice, induced immunogenic cell death and finally reduced the tumor burden, all this highlighting the therapeutic potential of this innovative approach. Overall, this study provides compelling evidence for the effectiveness of the INA of XVir-N-31 via shuttle cells as a promising therapeutic strategy for GBM. The non-invasive nature of the INA of OV-loaded shuttle cells holds great promise for future clinical translation. However, further research is required to assess the efficacy of this approach to ultimately progress in human clinical trials.

5.
J Clin Med ; 10(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204545

ABSTRACT

Amyloid-beta (Aß) deposition in the brain is the main pathological hallmark of Alzheimer disease. Peripheral clearance of Aß may possibly also lower brain levels. Recent evidence suggested that hepatic clearance of Aß42 is impaired in liver cirrhosis. To further test this hypothesis, serum Aß42 was measured by ELISA in portal venous serum (PVS), systemic venous serum (SVS), and hepatic venous serum (HVS) of 20 patients with liver cirrhosis. Mean Aß42 level was 24.7 ± 20.4 pg/mL in PVS, 21.2 ± 16.7 pg/mL in HVS, and 19.2 ± 11.7 pg/mL in SVS. Similar levels in the three blood compartments suggested that the cirrhotic liver does not clear Aß42. Aß42 was neither associated with the model of end-stage liver disease score nor the Child-Pugh score. Patients with abnormal creatinine or bilirubin levels or prolonged prothrombin time did not display higher Aß42 levels. Patients with massive ascites and patients with large varices had serum Aß42 levels similar to patients without these complications. Serum Aß42 was negatively associated with connective tissue growth factor levels (r = -0.580, p = 0.007) and a protective role of Aß42 in fibrogenesis was already described. Diabetic patients with liver cirrhosis had higher Aß42 levels (p = 0.069 for PVS, p = 0.047 for HVS and p = 0.181 for SVS), which is in accordance with previous reports. Present analysis showed that the cirrhotic liver does not eliminate Aß42. Further studies are needed to explore the association of liver cirrhosis, Aß42 levels, and cognitive dysfunction.

6.
Pharmaceuticals (Basel) ; 14(6)2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34199400

ABSTRACT

Noise is a wide-spread stress factor in modern life produced by urbanization, traffic, and an industrialized environment. Noise stress causes dysfunction and neurotransmission impairment in the central nervous system, as well as changes in hormone levels. In this study, we have examined the level of α-Tocopherol (α-T) and malondialdehyde (MDA) in plasma and the erythrocytes' membrane (EM), as well as the behavioral characteristics of a noise-induced stress model in rats. In addition, the modulating effect of α2-adrenoblockers, beditin, and mesedin on the aforementioned parameters has been investigated. For these purposes, albino male rats were divided into four groups: (1) untreated; (2) noise-exposed, (3) noise-exposed and beditin-treated (2 mg/kg, i.p.), and (4) noise-exposed and mesedin-treated (10 mg/kg, i.p.) animals. Noise-exposed groups were treated with 91dBA noise on 60 days with a daily duration of 8 h. Increased MDA and decreased α-T levels in plasma and EM were observed upon chronic high-level noise exposure. Locomotor and behavioral activity assessed with a Y-maze revealed disorientation and increased anxiety under chronic noise exposure. Prominently, α2-adrenoblockers alleviated both behavioral deficits and oxidative stress, providing evidence for the involvement of α2-adrenoceptor in the pathophysiology of noise-induced stress.

7.
Pharmaceuticals (Basel) ; 14(3)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809220

ABSTRACT

Huntington's disease (HD) is a monogenetic neurodegenerative disorder characterized by the accumulation of polyglutamine-expanded huntingtin (mHTT). There is currently no cure, and therefore disease-slowing remedies are sought to alleviate symptoms of the multifaceted disorder. Encouraging findings in Alzheimer's and Parkinson's disease on alpha-2 adrenoceptor (α2-AR) inhibition have shown neuroprotective and aggregation-reducing effects in cell and animal models. Here, we analyzed the effect of beditin, a novel α2- adrenoceptor (AR) antagonist, on cell viability and mHTT protein levels in cell models of HD using Western blot, time-resolved Foerster resonance energy transfer (TR-FRET), lactate dehydrogenase (LDH) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) cytotoxicity assays. Beditin decreases cytotoxicity, as measured by TUNEL staining and LDH release, in a neuronal progenitor cell model (STHdh cells) of HD and decreases the aggregation propensity of HTT exon 1 fragments in an overexpression model using human embryonic kidney (HEK) 293T cells. α2-AR is a promising therapeutic target for further characterization in HD models. Our data allow us to suggest beditin as a valuable candidate for the pharmaceutical manipulation of α2-AR, as it is capable of modulating neuronal cell survival and the level of mHTT.

8.
Pharmaceuticals (Basel) ; 14(2)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672482

ABSTRACT

Angiotensin receptor blockers (ARBs) have demonstrated multiple neuroprotective benefits in Alzheimer's disease (AD) models. However, their beneficial effects on memory deficits, cholinergic activity, neurogenesis and Amyloid beta (Aß) clearance reveal significant interstudy variability. The delivery route can impact not only delivery but also targeting and therapeutic efficacy of ARBs. Our previous findings on the beneficial effects of intranasally delivered losartan in the APP/PS1 model of AD prompted us to explore the influence of the delivery route by employing here the systemic administration of losartan. Consistent with our previous results with intranasal losartan, repeated intraperitoneal administration (10 mg/kg) resulted in a remarkable decrease in Aß plaques and soluble Aß42, as well as inflammatory cytokines (IL-2, IL-6 and TNFα). The Aß reduction can be ascribed to its facilitated degradation by neprilysin and diminished generation by BACE1. Losartan increased neurogenesis in vivo and in vitro and improved migratory properties of astrocytes isolated from adult transgenic AD mice. In summary, this data together with our previous results suggest therapeutic features of losartan which are independent of delivery route. The improvement of cell motility of Aß-affected astrocytes by losartan deserves further in vivo investigation, which may lead to new strategies for AD treatment.

9.
EBioMedicine ; 60: 102989, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32920368

ABSTRACT

BACKGROUND: Stem cells` (SC) functional heterogeneity and its poorly understood aetiology impedes clinical development of cell-based therapies in regenerative medicine and oncology. Recent studies suggest a strong correlation between the SC migration potential and their therapeutic efficacy in humans. Designating SC migration as a denominator of functional SC heterogeneity, we sought to identify highly migrating subpopulations within different SC classes and evaluate their therapeutic properties in comparison to the parental non-selected cells. METHODS: We selected highly migrating subpopulations from mesenchymal and neural SC (sMSC and sNSC), characterized their features including but not limited to migratory potential, trophic factor release and transcriptomic signature. To assess lesion-targeted migration and therapeutic properties of isolated subpopulations in vivo, surgical transplantation and intranasal administration of MSCs in mouse models of glioblastoma and Alzheimer's disease respectively were performed. FINDINGS: Comparison of parental non-selected cells with isolated subpopulations revealed superior motility and migratory potential of sMSC and sNSC in vitro. We identified podoplanin as a major regulator of migratory features of sMSC/sNSC. Podoplanin engineering improved oncovirolytic activity of virus-loaded NSC on distantly located glioblastoma cells. Finally, sMSC displayed more targeted migration to the tumour site in a mouse glioblastoma model and remarkably higher potency to reduce pathological hallmarks and memory deficits in transgenic Alzheimer's disease mice. INTERPRETATION: Functional heterogeneity of SC is associated with their motility and migration potential which can serve as predictors of SC therapeutic efficacy. FUNDING: This work was supported in part by the Robert Bosch Stiftung (Stuttgart, Germany) and by the IZEPHA grant.


Subject(s)
Cell Movement , Stem Cells/physiology , Alzheimer Disease/therapy , Animals , Biomarkers , Cell Survival , Cell Tracking/methods , Cells, Cultured , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Humans , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Oncolytic Virotherapy , Stem Cell Transplantation , Stem Cells/cytology , Treatment Outcome
10.
EBioMedicine ; 60: 102987, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32942121

ABSTRACT

BACKGROUND: Limited knowledge of stem cell therapies` mechanisms of action hampers their sustainable implementation into the clinic. Specifically, the interactions of transplanted stem cells with the host vasculature and its implications for their therapeutic efficacy are not elucidated. We tested whether adhesion receptors and chemokine receptors on stem cells can be functionally modulated, and consequently if such modulation may substantially affect therapeutically relevant stem cell interactions with the host endothelium. METHODS: We investigated the effects of cationic molecule polyethylenimine (PEI) treatment with or without nanoparticles on the functions of adhesion receptors and chemokine receptors of human bone marrow-derived Mesenchymal Stem Cells (MSC). Analyses included MSC functions in vitro, as well as homing and therapeutic efficacy in rodent models of central nervous system´s pathologies in vivo. FINDINGS: PEI treatment did not affect viability, immunomodulation or differentiation potential of MSC, but increased the CCR4 expression and functionally blocked their adhesion receptors, thus decreasing their adhesion capacity in vitro. Intravenously applied in a rat model of brain injury, the homing rate of PEI-MSC in the brain was highly increased with decreased numbers of adherent PEI-MSC in the lung vasculature. Moreover, in comparison to untreated MSC, PEI-MSC featured increased tumour directed migration in a mouse glioblastoma model, and superior therapeutic efficacy in a murine model of stroke. INTERPRETATION: Balanced stem cell adhesion and migration in different parts of the vasculature and tissues together with the local microenvironment impacts their therapeutic efficacy. FUNDING: Robert Bosch Stiftung, IZEPHA grant, EU grant 7 FP Health.


Subject(s)
Cell Adhesion , Cell Movement , Endothelium/metabolism , Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line , Cell- and Tissue-Based Therapy , Cells, Cultured , Cellular Microenvironment , Disease Models, Animal , Glioma/diagnosis , Glioma/pathology , Glioma/therapy , Humans , Immunophenotyping , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Rats , Stem Cell Transplantation , Xenograft Model Antitumor Assays
11.
Cells ; 9(2)2020 02 17.
Article in English | MEDLINE | ID: mdl-32089540

ABSTRACT

The function and regulation of amyloid-beta (Aß) in healthy and diseased liver remains unexplored. Because Aß reduces the integrity of the blood-brain barrier we have examined its potential role in regulating the sinusoidal permeability of normal and cirrhotic liver. Aß and key proteins that generate (beta-secretase 1 and presenilin-1) and degrade it (neprilysin and myelin basic protein) were decreased in human cirrhotic liver. In culture, activated hepatic stellate cells (HSC) internalized Aß more efficiently than astrocytes and HSC degraded Aß leading to suppressed expression of α-smooth muscle actin (α-SMA), collagen 1 and transforming growth factor ß (TGFß). Aß also upregulated sinusoidal permeability marker endothelial NO synthase (eNOS) and decreased TGFß in cultured human liver sinusoidal endothelial cells (hLSEC). Liver Aß levels also correlate with the expression of eNOS in transgenic Alzheimer's disease mice and in human and rodent cirrhosis/fibrosis. These findings suggest a previously unexplored role of Aß in the maintenance of liver sinusoidal permeability and in protection against cirrhosis/fibrosis via attenuation of HSC activation.


Subject(s)
Amyloid beta-Peptides/therapeutic use , Fibrosis/drug therapy , Gene Expression/genetics , Liver Cirrhosis/therapy , Peptide Fragments/therapeutic use , Amyloid beta-Peptides/pharmacology , Animals , Disease Models, Animal , Humans , Liver Cirrhosis/physiopathology , Male , Mice , Mice, Transgenic , Middle Aged , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley
12.
Cells ; 9(2)2020 02 18.
Article in English | MEDLINE | ID: mdl-32085378

ABSTRACT

The noradrenergic system is proposed to play a prominent role in the pathogenesis of liver fibrosis. While α1- and ß-adrenergic receptors (ARs) are suggested to be involved in a multitude of profibrogenic actions, little is known about α2-AR-mediated effects and their expression pattern during liver fibrosis and cirrhosis. We explored the expression of α2-AR in two models of experimental liver fibrosis. We further evaluated the capacity of the α2-AR blocker mesedin to deactivate hepatic stellate cells (HSCs) and to increase the permeability of human liver sinusoidal endothelial cells (hLSECs). The mRNA of α2a-, α2b-, and α2c-AR subtypes was uniformly upregulated in carbon tetrachloride-treated mice vs the controls, while in bile duct-ligated mice, only α2b-AR increased in response to liver injury. In murine HSCs, mesedin led to a decrease in α-smooth muscle actin, transforming growth factor-ß and α2a-AR expression, which was indicated by RT-qPCR, immunocytochemistry, and Western blot analyses. In a hLSEC line, an increased expression of endothelial nitric oxide synthase was detected along with downregulated transforming growth factor-ß. In conclusion, we suggest that the α2-AR blockade alleviates the activation of HSCs and may increase the permeability of liver sinusoids during liver injury.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Dioxanes/therapeutic use , Liver Cirrhosis/drug therapy , Receptors, Adrenergic, alpha-2/genetics , Thiazoles/therapeutic use , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Dioxanes/pharmacology , Disease Models, Animal , Female , Humans , Liver Cirrhosis/physiopathology , Mice , Thiazoles/pharmacology
13.
Cells ; 8(6)2019 06 15.
Article in English | MEDLINE | ID: mdl-31208073

ABSTRACT

Intrastriatal administration of mesenchymal stem cells (MSCs) has shown beneficial effects in rodent models of Huntington disease (HD). However, the invasive nature of surgical procedure and its potential to trigger the host immune response may limit its clinical use. Hence, we sought to evaluate the non-invasive intranasal administration (INA) of MSC delivery as an effective alternative route in HD. GFP-expressing MSCs derived from bone marrow were intranasally administered to 4-week-old R6/2 HD transgenic mice. MSCs were detected in the olfactory bulb, midbrain and striatum five days post-delivery. Compared to phosphate-buffered saline (PBS)-treated littermates, MSC-treated R6/2 mice showed an increased survival rate and attenuated circadian activity disruption assessed by locomotor activity. MSCs increased the protein expression of DARPP-32 and tyrosine hydroxylase (TH) and downregulated gene expression of inflammatory modulators in the brain 7.5 weeks after INA. While vehicle treated R6/2 mice displayed decreased Iba1 expression and altered microglial morphology in comparison to the wild type littermates, MSCs restored both, Iba1 level and the thickness of microglial processes in the striatum of R6/2 mice. Our results demonstrate significantly ameliorated phenotypes of R6/2 mice after MSCs administration via INA, suggesting this method as an effective delivering route of cells to the brain for HD therapy.


Subject(s)
Dopamine/metabolism , Huntington Disease/physiopathology , Huntington Disease/therapy , Inflammation/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Synaptic Transmission , Administration, Intranasal , Animals , Brain/pathology , Brain/physiopathology , Cell Tracking , Circadian Rhythm , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation , Humans , Huntington Disease/genetics , Inflammation/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Motor Activity , Nerve Growth Factors/metabolism , Sleep , Survival Analysis , Tyrosine 3-Monooxygenase/metabolism
14.
Neurotherapeutics ; 16(3): 725-740, 2019 07.
Article in English | MEDLINE | ID: mdl-30796737

ABSTRACT

The contribution of the local angiotensin receptor system to neuroinflammation, impaired neurogenesis, and amyloid beta (Aß) accumulation in Alzheimer's disease (AD) and in hypertension is consistent with the remarkable neuroprotection provided by angiotensin receptor blockers (ARBs) independent of their blood pressure-lowering effect. Considering the causal relationship between hypertension and AD and that targeting cerebrovascular pathology with ARBs does not necessarily require their systemic effects, we tested intranasal losartan in the rat model of chronic hypertension (spontaneously hypertensive stroke-prone rats, SHRSP). Intranasal losartan at a subdepressor dose decreased mortality, neuroinflammation, and perivascular content of Aß by enhancing key players in its metabolism and clearance, including insulin-degrading enzyme, neprilysin, and transthyretin. Furthermore, this treatment improved neurologic deficits and increased brain IL-10 concentration, hippocampal cell survival, neurogenesis, and choroid plexus cell proliferation in SHRSP. Losartan (1 µM) also reduced LDH release from cultured astroglial cells in response to toxic glutamate concentrations. This effect was completely blunted by IL-10 antibodies. These findings suggest that intranasal ARB treatment is a neuroprotective, neurogenesis-inducing, and Aß-decreasing strategy for the treatment of hypertensive stroke and cerebral amyloid angiopathy acting at least partly through the IL-10 pathway.


Subject(s)
Amyloid beta-Peptides/metabolism , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Glymphatic System/chemistry , Hypertension/complications , Inflammation/drug therapy , Losartan/therapeutic use , Neurogenesis/drug effects , Stroke/prevention & control , Administration, Intranasal , Animals , Dose-Response Relationship, Drug , Losartan/administration & dosage , Male , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Stroke/etiology
15.
Int J Mol Sci ; 19(1)2017 Dec 21.
Article in English | MEDLINE | ID: mdl-29267189

ABSTRACT

Locus coeruleus-noradrenergic system dysfunction is known to contribute to the progression of Alzheimer's disease (AD). Besides a variety of reports showing the involvement of norepinephrine and its receptor systems in cognition, amyloid ß (Aß) metabolism, neuroinflammation, and neurogenesis, little is known about the contribution of the specific receptors to these actions. Here, we investigated the neurogenic and neuroprotective properties of a new α2 adrenoblocker, mesedin, in astroglial primary cultures (APC) from C57BL/6 and 3×Tg-AD mice. Our results demonstrate that mesedin rescues neuronal precursors and young neurons, and reduces the lactate dehydrogenase (LDH) release from astroglia under hypoxic and normoxic conditions. Mesedin also increased choline acetyltransferase, postsynaptic density marker 95 (PSD95), and Aß-degrading enzyme neprilysin in the wild type APC, while in the 3×Tg-AD APC exposed to glutamate, it decreased the intracellular content of Aß and enhanced the survival of synaptophysin-positive astroglia and neurons. These effects in APC can at least partially be attributed to the mesedin's ability of increasing the expression of Interleukine(IL)-10, which is a potent anti-inflammatory, neuroprotective neurogenic, and Aß metabolism enhancing factor. In summary, our data identify the neurogenic, neuroprotective, and anti-amyloidogenic action of mesedin in APC. Further in vivo studies are needed to estimate the therapeutic value of mesedin for AD.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Astrocytes/drug effects , Dioxanes/pharmacology , Dioxanes/therapeutic use , Neurogenesis/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Thiazoles/pharmacology , Thiazoles/therapeutic use , Adrenergic alpha-2 Receptor Antagonists/chemistry , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Alzheimer Disease/drug therapy , Animals , Astrocytes/cytology , Biomarkers, Pharmacological/analysis , Cell Survival/drug effects , Dioxanes/chemistry , Glutamic Acid/metabolism , Hypoxia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Thiazoles/chemistry
16.
Health Qual Life Outcomes ; 15(1): 171, 2017 Aug 25.
Article in English | MEDLINE | ID: mdl-28841891

ABSTRACT

BACKGROUND: A number of the QL researches in case of different pathologies are being increased during the last decade. The existing traditional research methods provide mostly arbitrary data on the disease and its treatment, which are not sufficient for the schoolchildren overall psychological and social adaptation and wellness evaluation. METHODS: The research object became schoolchildren of 3 randomly selected schools in Yerevan. 443 monitoring units formed the selection population. The degree of situational and personal anxiety was evaluated with the help of Spielberger's and Gerbachevski's tests. RESULTS: According to our research data the anxiety degree was 29,2 ± 2,3 points among the girls and 12,5 ± 1,6 points among the boys, respectively. The individual anxiety level was especially high: it made up 44,5 ± 0,8 points, and that of the situational anxiety made up 37,2 ± 0,5 points (p < 0,05). According to Gerbachovski's test in the group of schoolchildren with ENT pathology those with a high level of demands made up 53,5 ± 3,2%, with a medium level of demands - 32,4 ± 3,0% and with a low level of demands -14,1 ± 2,2%. A number of the practically healthy schoolchildren with a low level of demands made up 50,3%, and with a high level - 30,7%. CONCLUSION: According to the investigation data those children who suffer from the ENT chronic diseases usually avoided communication, were sluggish and shy. According to the results of the research, the socio-psychological and adaptation abilities of children with the ENT chronic diseases were lower than those of the practically healthy (without ENT pathologies) coevals. This fact urges to improve the prophylactic measures provision in the mentioned pathologies aspect.


Subject(s)
Anxiety/psychology , Otorhinolaryngologic Diseases/psychology , Quality of Life , Social Adjustment , Stress, Psychological/psychology , Adolescent , Case-Control Studies , Child , Chronic Disease/psychology , Cross-Sectional Studies , Female , Humans , Male , Parents/psychology , Sex Factors , Surveys and Questionnaires
17.
Cell Transplant ; 23 Suppl 1: S123-39, 2014.
Article in English | MEDLINE | ID: mdl-25302802

ABSTRACT

In view of the rapid preclinical development of cell-based therapies for neurodegenerative disorders, traumatic brain injury, and tumors, the safe and efficient delivery and targeting of therapeutic cells to the central nervous system is critical for maintaining therapeutic efficacy and safety in the respective disease models. Our previous data demonstrated therapeutically efficacious and targeted delivery of mesenchymal stem cells (MSCs) to the brain in the rat 6-hydroxydopamine model of Parkinson's disease (PD). The present study examined delivery of bone marrow-derived MSCs, macrophages, and microglia to the brain in a transgenic model of PD [(Thy1)-h[A30P] αS] and an APP/PS1 model of Alzheimer's disease (AD) via intranasal application (INA). INA of microglia in naive BL/6 mice led to targeted and effective delivery of cells to the brain. Quantitative PCR analysis of eGFP DNA showed that the brain contained the highest amount of eGFP-microglia (up to 2.1 × 10(4)) after INA of 1 × 10(6) cells, while the total amount of cells detected in peripheral organs did not exceed 3.4 × 10(3). Seven days after INA, MSCs expressing eGFP were detected in the olfactory bulb (OB), cortex, amygdala, striatum, hippocampus, cerebellum, and brainstem of (Thy1)-h[A30P] αS transgenic mice, showing predominant distribution within the OB and brainstem. INA of eGFP-expressing macrophages in 13-month-old APP/PS1 mice led to delivery of cells to the OB, hippocampus, cortex, and cerebellum. Both MSCs and macrophages contained Iba-1-positive population of small microglia-like cells and Iba-1-negative large rounded cells showing either intracellular amyloid ß (macrophages in APP/PS1 model) or α-synuclein [MSCs in (Thy1)-h[A30P] αS model] immunoreactivity. Here, we show, for the first time, intranasal delivery of cells to the brain of transgenic PD and AD mouse models. Additional work is needed to determine the optimal dosage (single treatment regimen or repeated administrations) to achieve functional improvement in these mouse models with intranasal microglia/macrophages and MSCs. This manuscript is published as part of the International Association of Neurorestoratology (IANR) special issue of Cell Transplantation.


Subject(s)
Alzheimer Disease/therapy , Brain/pathology , Macrophages/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Microglia/transplantation , Parkinson Disease/therapy , Administration, Intranasal , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cell Lineage , Disease Models, Animal , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Int J Mol Sci ; 15(2): 2858-75, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24557580

ABSTRACT

We show that, under in vitro conditions, the vulnerability of astroglia to hypoxia is reflected by alterations in endothelin (ET)-1 release and capacity of erythropoietin (EPO) to regulate ET-1 levels. Exposure of cells to 24 h hypoxia did not induce changes in ET-1 release, while 48-72 h hypoxia resulted in increase of ET-1 release from astrocytes that could be abolished by EPO. The endothelin receptor type A (ETA) antagonist BQ123 increased extracellular levels of ET-1 in human fetal astroglial cell line (SV-FHAS). The survival and proliferation of rat primary astrocytes, neural precursors, and neurons upon hypoxic conditions were increased upon administration of BQ123. Hypoxic injury and aging affected the interaction between the EPO and ET systems. Under hypoxia EPO decreased ET-1 release from astrocytes, while ETA receptor blockade enhanced the expression of EPO mRNA and EPO receptor in culture-aged rat astroglia. The blockade of ETA receptor can increase the availability of ET-1 to the ETB receptor and can potentiate the neuroprotective effects of EPO. Thus, the new therapeutic use of combined administration of EPO and ETA receptor antagonists during hypoxia-associated neurodegenerative disorders of the central nervous system (CNS) can be suggested.


Subject(s)
Cell Hypoxia , Endothelin-1/metabolism , Erythropoietin/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Erythropoietin/genetics , Humans , Peptides, Cyclic/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Endothelin A/agonists , Receptor, Endothelin A/metabolism , Receptors, Erythropoietin/metabolism , Up-Regulation/drug effects
19.
PLoS One ; 8(10): e77182, 2013.
Article in English | MEDLINE | ID: mdl-24124607

ABSTRACT

Extracellular accumulation of toxic concentrations of glutamate (Glu) is a hallmark of many neurodegenerative diseases, often accompanied by hypoxia and impaired metabolism of this neuromediator. To address the question whether the multifunctional neuroprotective action of erythropoietin (EPO) extends to the regulation of extracellular Glu-level and is age-related, young and culture-aged rat astroglial primary cells (APC) were simultaneously treated with 1mM Glu and/or human recombinant EPO under normoxic and hypoxic conditions (NC and HC). EPO increased the Glu uptake by astrocytes under both NC and especially upon HC in culture-aged APC (by 60%). Moreover, treatment with EPO up-regulated the activity of glutamine synthetase (GS), the expression of glutamate-aspartate transporter (GLAST) and the level of EPO mRNA. EPO alleviated the Glu- and hypoxia-induced LDH release from astrocytes. These protective EPO effects were concentration-dependent and they were strongly intensified with age in culture. More than a 4-fold increase in apoptosis and a 2-fold decrease in GS enzyme activity was observed in APC transfected with EPO receptor (EPOR)-siRNA. Our in vivo data show decreased expression of EPO and a strong increase of EPOR in brain homogenates of APP/PS1 mice and their wild type controls during aging. Comparison of APP/PS1 and age-matched WT control mice revealed a stronger expression of EPOR but a weaker one of EPO in the Alzheimer's disease (AD) model mice. Here we show for the first time the direct correlation between the extent of differentiation (age) of astrocytes and the efficacy of EPO in balancing extracellular glutamate clearance and metabolism in an in-vitro model of hypoxia and Glu-induced astroglial injury. The clinical relevance of EPO and EPOR as markers of brain cells vulnerability during aging and neurodegeneration is evidenced by remarkable changes in their expression levels in a transgenic model of AD and their WT controls.


Subject(s)
Astrocytes/metabolism , Erythropoietin/metabolism , Glutamic Acid/metabolism , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Astrocytes/drug effects , Brain/metabolism , Cell Hypoxia , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cellular Senescence , Enzyme Activation/drug effects , Erythropoietin/genetics , Erythropoietin/pharmacology , Extracellular Space/metabolism , Gene Expression Regulation/drug effects , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/drug effects , Male , Rats , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism
20.
Pharm Res ; 30(10): 2475-84, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23135822

ABSTRACT

One of the most challenging problems facing modern medicine is how to deliver a given drug to a specific target at the exclusion of other regions. For example, a variety of compounds have beneficial effects within the central nervous system (CNS), but unwanted side effects in the periphery. For such compounds, traditional oral or intravenous drug delivery fails to provide benefit without cost. However, intranasal delivery is emerging as a noninvasive option for delivering drugs to the CNS with minimal peripheral exposure. Additionally, this method facilitates the delivery of large and/or charged therapeutics, which fail to effectively cross the blood-brain barrier (BBB). Thus, for a variety of growth factors, hormones, neuropeptides and therapeutics including insulin, oxytocin, orexin, and even stem cells, intranasal delivery is emerging as an efficient method of administration, and represents a promising therapeutic strategy for the treatment of diseases with CNS involvement, such as obesity, Alzheimer's disease, Parkinson's disease, Huntington's disease, depression, anxiety, autism spectrum disorders, seizures, drug addiction, eating disorders, and stroke.


Subject(s)
Central Nervous System Diseases/drug therapy , Drug Delivery Systems/methods , Pharmaceutical Preparations/administration & dosage , Administration, Intranasal , Animals , Central Nervous System Diseases/metabolism , Drug Delivery Systems/adverse effects , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...