Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731928

ABSTRACT

Neurodegenerative disorders (NDs) and the development of various therapeutic strategies to combat them have received increased attention in recent decades [...].


Subject(s)
Neurodegenerative Diseases , Neurodegenerative Diseases/therapy , Neurodegenerative Diseases/metabolism , Humans , Animals
2.
Biol Reprod ; 110(1): 63-77, 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-37741056

ABSTRACT

Little is known about the non-neuronal spermic cholinergic system, which may regulate sperm motility and the acrosome reaction initiation process. We investigated the presence of the key acetylcholine (ACh)-biosynthesizing enzyme, choline acetyltransferase (ChAT), and the acetylcholine-degrading enzymes, acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) and two ACh-receptors in human spermatozoa and seminal plasma. Fresh ejaculates were used for intra- and extracellular flow cytometric analysis of ChAT, AChE, BChE, and alpha-7-nicotinic and M1-muscarinic ACh-receptors in sperm. For determining the source of soluble enzymes, frozen seminal samples (n = 74) were selected on two bases: (1) from vasectomized (n = 37) and non-vasectomized (n = 37) subjects and (2) based on levels of alpha-glucosidase, fructose, or zinc to define sample subgroups with high or low fluid contribution from the epididymis and seminal vesicle, and prostate, respectively. Flow cytometric analyses revealed that ChAT was expressed intracellularly in essentially all spermatozoa. ChAT was also present in a readily membrane-detachable form at the extracellular membrane of at least 18% of the spermatozoa. These were also highly positive for intra- and extracellular BChE (>83%) and M1 (>84%) and α7 (>59%) ACh-receptors. Intriguingly, the sperm was negative for AChE. Analyses of seminal plasma revealed that spermatozoa and epididymides were major sources of soluble ChAT and BChE, whereas soluble AChE most likely originated from epididymides and seminal vesicles. Prostate had relatively minor contribution to the pool of the soluble enzymes in the seminal fluid. In conclusion, human spermatozoa exhibited a cholinergic phenotype and were one of the major sources of soluble ChAT and BChE in ejaculate. We also provide the first evidence for ChAT as an extracellularly membrane-anchored protein.


Subject(s)
Acetylcholine , Acetylcholinesterase , Humans , Male , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Semen/metabolism , Sperm Motility , Spermatozoa/metabolism , Cholinergic Agents
3.
Alzheimers Res Ther ; 15(1): 137, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37596686

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is an age-related disease characterized by altered cognition, neuroinflammation, and neurodegeneration against which there is presently no effective cure. Brain-derived neurotrophic factor (BDNF) is a key neurotrophin involved in the learning and memory process, with a crucial role in synaptic plasticity and neuronal survival. Several findings support that a reduced BDNF expression in the human brain is associated with AD pathogenesis. BDNF has been proposed as a potential therapy for AD, but BDNF has low brain penetration. In this study, we used an innovative encapsulated cell biodelivery (ECB) device, containing genetically modified cells capable of releasing BDNF and characterized its feasibility and therapeutic effects in the novel App knock-in AD mouse model (AppNL-G-F). METHODS: ECB's containing human ARPE-19 cells genetically modified to release BDNF (ECB-BDNF devices) were stereotactically implanted bilaterally into hippocampus of 3-month-old AppNL-G-F mice. The stability of BDNF release and its effect on AD pathology were evaluated after 1, 2-, and 4-months post-implantation by immunohistochemical and biochemical analyses. Exploratory and memory performance using elevated plus maze (EPM) and Y-maze test were performed in the 4-months treatment group. Immunological reaction towards ECB-BDNF devices were studied under ex vivo and in vivo settings. RESULTS: The surgery and the ECB-BDNF implants were well tolerated without any signs of unwanted side effects or weight loss. ECB-BDNF devices did not induce host-mediated immune response under ex vivo set-up but showed reduced immune cell attachment when explanted 4-months post-implantation. Elevated BDNF staining around ECB-BDNF device proximity was detected after 1, 2, and 4 months treatment, but the retrieved devices showed variable BDNF release. A reduction of amyloid-ß (Aß) plaque deposition was observed around ECB-BDNF device proximity after 2-months of BDNF delivery. CONCLUSIONS: The result of this study supports the use of ECB device as a promising drug-delivery approach to locally administer BBB-impermeable factors for treating neurodegenerative conditions like AD. Optimization of the mouse-sized devices to reduce variability of BDNF release is needed to employ the ECB platform in future pre-clinical research and therapy development studies.


Subject(s)
Alzheimer Disease , Brain-Derived Neurotrophic Factor , Drug Delivery Systems , Animals , Mice , Alzheimer Disease/therapy , Amyloid beta-Peptides , Brain-Derived Neurotrophic Factor/therapeutic use , Feasibility Studies , Drug Delivery Systems/methods
4.
Acta Neuropathol Commun ; 11(1): 76, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37158955

ABSTRACT

Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by NOTCH3 mutations. Typical CADASIL is characterised by subcortical ischemic strokes due to severe arteriopathy and fibrotic thickening of small arteries. Arteriolar vascular smooth muscle cells (VSMCs) are the key target in CADASIL, but the potential mechanisms involved in their degeneration are still unclear. Focusing on cerebral microvessels in the frontal and anterior temporal lobes and the basal ganglia, we used advanced proteomic and immunohistochemical methods to explore the extent of inflammatory and immune responses in CADASIL subjects compared to similar age normal and other disease controls. There was variable loss of VSMC in medial layers of arteries in white matter as well as the cortex, that could not be distinguished whether NOTCH3 mutations were in the epidermal growth factor (EGFr) domains 1-6 or EGFr7-34. Proteomics of isolated cerebral microvessels showed alterations in several proteins, many associated with endoplasmic reticulum (ER) stress including heat shock proteins. Cerebral vessels with sparsely populated VSMCs also attracted robust accrual of perivascular microglia/macrophages in order CD45+ > CD163+ > CD68+cells, with > 60% of vessel walls exhibiting intercellular adhesion molecule-1 (ICAM-1) immunoreactivity. Functional VSMC cultures bearing the NOTCH3 Arg133Cys mutation showed increased gene expression of the pro-inflammatory cytokine interleukin 6 and ICAM-1 by 16- and 50-fold, respectively. We further found evidence for activation of the alternative pathway of complement. Immunolocalisation of complement Factor B, C3d and C5-9 terminal complex but not C1q was apparent in ~ 70% of cerebral vessels. Increased complement expression was corroborated in > 70% of cultured VSMCs bearing the Arg133Cys mutation independent of N3ECD immunoreactivity. Our observations suggest that ER stress and other cellular features associated with arteriolar VSMC damage instigate robust localized inflammatory and immune responses in CADASIL. Our study has important implications for immunomodulation approaches to counter the characteristic arteriopathy of CADASIL.


Subject(s)
CADASIL , Humans , CADASIL/genetics , Intercellular Adhesion Molecule-1 , Proteomics , Complement System Proteins , Cerebral Infarction
5.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012296

ABSTRACT

There is no cure yet available for Alzheimer's disease (AD). We recently optimized encapsulated cell biodelivery (ECB) devices releasing human mature nerve growth factor (hmNGF), termed ECB-NGF, to the basal forebrain of AD patients. The ECB-NGF delivery resulted in increased CSF cholinergic markers, improved glucose metabolism, and positive effects on cognition in AD patients. However, some ECB-NGF implants showed altered hmNGF release post-explantation. To optimize the ECB-NGF platform for future therapeutic purposes, we initiated in-vitro optimization studies by exposing ECB-NGF devices to physiological factors present within the AD brain. We report here that microglia cells can impair hmNGF release from ECB-NGF devices in-vitro, which can be reversed by transferring the devices to fresh culture medium. Further, we exposed the hmNGF secreting human ARPE-19 cell line (NGC0211) to microglia (HMC3) conditioned medium (MCM; untreated or treated with IL-1ß/IFNγ/Aß40/Aß42), and evaluated biochemical stress markers (ROS, GSH, ΔΨm, and Alamar Blue assay), cell death indicators (Annexin-V/PI), cell proliferation (CFSE retention and Ki67) and senescence markers (SA-ß-gal) in NGC0211 cells. MCMs from activated microglia reduced cell proliferation and induced cell senescence in NGC0211 cells, which otherwise resist biochemical alterations and cell death. These data indicate a critical but reversible impact of activated microglia on NGC0211 cells.


Subject(s)
Alzheimer Disease , Basal Forebrain , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Basal Forebrain/metabolism , Biomarkers , Cell Proliferation , Humans , Microglia/metabolism , Nerve Growth Factor/metabolism
6.
EBioMedicine ; 77: 103882, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35182996

ABSTRACT

BACKGROUND: Post-traumatic syringomyelia (PTS) affects patients with chronic spinal cord injury (SCI) and is characterized by progressive deterioration of neurological symptoms. To improve surgical treatment, we studied the therapeutic effects of neuroepithelial-like stem cells (NESCs) derived from induced pluripotent stem cells (iPSCs) in a rat model of PTS. To facilitate clinical translation, we studied NESCs derived from Good Manufacturing Practice (GMP)-compliant iPSCs. METHODS: Human GMP-compliant iPSCs were used to derive NESCs. Cryo-preserved NESCs were used off-the-shelf for intraspinal implantation to PTS rats 1 or 10 weeks post-injury, and rats were sacrificed 10 weeks later. In vivo cyst volumes were measured with micro-MRI. Phenotypes of differentiated NESCs and host responses were analyzed by immunohistochemistry. FINDINGS: Off-the-shelf NESCs transplanted to PTS rats 10 weeks post-injury reduced cyst volume. The grafted NESCs differentiated mainly into glial cells. Importantly, NESCs also stimulated tissue repair. They reduced the density of glial scars and neurite-inhibiting chondroitin sulfate proteoglycan 4 (CSPG4), stimulated host oligodendrocyte precursor cells to migrate and proliferate, reduced active microglia/macrophages, and promoted axonal regrowth after subacute as well as chronic transplantation. INTERPRETATION: Significant neural repair promoted by NESCs demonstrated that human NESCs could be used as a complement to standard surgery in PTS. We envisage that future PTS patients transplanted with NESCs will benefit both from eliminating the symptoms of PTS, as well as a long-term improvement of the neurological symptoms of SCI. FUNDING: This work was supported by Vinnova (2016-04134), Karolinska Institutet StratRegen, and the Chinese Scholarship Council.


Subject(s)
Induced Pluripotent Stem Cells , Neural Stem Cells , Spinal Cord Injuries , Syringomyelia , Animals , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/transplantation , Rats , Spinal Cord Injuries/complications , Spinal Cord Injuries/therapy , Syringomyelia/etiology , Syringomyelia/therapy
7.
Cells ; 10(11)2021 10 21.
Article in English | MEDLINE | ID: mdl-34831056

ABSTRACT

Alzheimer's disease (AD) treatment is constrained due to the inability of peripherally administered therapeutic molecules to cross the blood-brain barrier. Encapsulated cell biodelivery (ECB) devices, a tissue-targeted approach for local drug release, was previously optimized for human mature nerve growth factor (hmNGF) delivery in AD patients but was found to have reduced hmNGF release over time. To understand the reason behind reduced ECB efficacy, we exposed hmNGF-releasing cells (NGC0211) in vitro to human cerebrospinal fluid (CSF) obtained from Subjective Cognitive Impairment (SCI), Lewy Body Dementia (LBD), and AD patients. Subsequently, we exposed NGC0211 cells directly to AD-related factors like amyloid-ß peptides (Aß40/42) or activated astrocyte-conditioned medium (Aß40/42/IL-1ß/TNFα-treated) and evaluated biochemical stress markers, cell death indicators, cell proliferation marker (Ki67), and hmNGF release. We found that all patients' CSF significantly reduced hmNGF release from NGC0211 cells in vitro. Aß40/42, inflammatory molecules, and activated astrocytes significantly affected NGC0211 cell proliferation without altering hmNGF release or other parameters important for essential functions of the NGC0211 cells. Long-term constant cell proliferation within the ECB device is critically important to maintain a steady cell population needed for stable mNGF release. These data show hampered proliferation of NGC0211 cells, which may lead to a decline of the NGC0211 cell population in ECBs, thereby reducing hmNGF release. Our study highlights the need for future studies to strengthen ECB-mediated long-term drug delivery approaches.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Cells, Immobilized/cytology , Nerve Growth Factor/metabolism , Alzheimer Disease/cerebrospinal fluid , Cell Line , Cell Proliferation , Cognitive Dysfunction/cerebrospinal fluid , Culture Media, Conditioned/pharmacology , Humans , Lewy Body Disease/cerebrospinal fluid , Peptides/metabolism , Stress, Physiological
8.
Front Aging Neurosci ; 13: 714186, 2021.
Article in English | MEDLINE | ID: mdl-34475820

ABSTRACT

Gradual decline in cholinergic transmission and cognitive function occurs during normal aging, whereas pathological loss of cholinergic function is a hallmark of different types of dementia, including Alzheimer's disease (AD), Lewy body dementia (LBD), and Parkinson's disease dementia (PDD). Glial cell line-derived neurotrophic factor (GDNF) is known to modulate and enhance the dopamine system. However, how endogenous GDNF influences brain cholinergic transmission has remained elusive. In this study, we explored the effect of a twofold increase in endogenous GDNF (Gdnf hypermorphic mice, Gdnf wt/hyper) on cholinergic markers and cognitive function upon aging. We found that Gdnf wt/hyper mice resisted an overall age-associated decline in the cholinergic index observed in the brain of Gdnf wt/wt animals. Biochemical analysis revealed that the level of nerve growth factor (NGF), which is important for survival and function of central cholinergic neurons, was significantly increased in several brain areas of old Gdnf wt/hyper mice. Analysis of expression of genes involved in cholinergic transmission in the cortex and striatum confirmed modulation of cholinergic pathways by GDNF upon aging. In line with these findings, Gdnf wt/hyper mice did not undergo an age-related decline in cognitive function in the Y-maze test, as observed in the wild type littermates. Our results identify endogenous GDNF as a potential modulator of cholinergic transmission and call for future studies on endogenous GDNF function in neurodegenerative disorders characterized by cognitive impairments, including AD, LBD, and PDD.

9.
Adv Exp Med Biol ; 1331: 167-191, 2021.
Article in English | MEDLINE | ID: mdl-34453298

ABSTRACT

Age-dependent progressive neurodegeneration and associated cognitive dysfunction represent a serious concern worldwide. Currently, dementia accounts for the fifth highest cause of death, among which Alzheimer's disease (AD) represents more than 60% of the cases. AD is associated with progressive cognitive dysfunction which affects daily life of the affected individual and associated family. The cognitive dysfunctions are at least partially due to the degeneration of a specific set of neurons (cholinergic neurons) whose cell bodies are situated in the basal forebrain region (basal forebrain cholinergic neurons, BFCNs) but innervate wide areas of the brain. It has been explicitly shown that the delivery of the neurotrophic protein nerve growth factor (NGF) can rescue BFCNs and restore cognitive dysfunction, making NGF interesting as a potential therapeutic substance for AD. Unfortunately, NGF cannot pass through the blood-brain barrier (BBB) and thus peripheral administration of NGF protein is not viable therapeutically. NGF must be delivered in a way which will allow its brain penetration and availability to the BFCNs to modulate BFCN activity and viability. Over the past few decades, various methodologies have been developed to deliver NGF to the brain tissue. In this chapter, NGF delivery methods are discussed in the context of AD.


Subject(s)
Alzheimer Disease , Basal Forebrain , Alzheimer Disease/drug therapy , Humans , Nerve Growth Factor/metabolism , Neurons/metabolism
10.
Handb Clin Neurol ; 179: 219-229, 2021.
Article in English | MEDLINE | ID: mdl-34225964

ABSTRACT

There is currently no effective treatment for the most common of the dementia disorders, Alzheimer's disease (AD). It has been known for decades that the central cholinergic system is important for memory. The cholinergic neurons in the basal forebrain with its cortical and hippocampal projections degenerate in AD and thus contribute to the cognitive decline characteristic of AD. This knowledge led to the development of the currently approved treatment for AD, with inhibitors of acetylcholine-esterase targeting the cholinergic system with beneficial but mild effects. In recent years, other approaches to influence the degenerating cholinergic system in AD focusing on nerve growth factor (NGF) have been undertaken. NGF is required for the survival and function of the basal forebrain cholinergic neurons, the most important being the nucleus basalis of Meynert (nbM). Since there is a lack of NGF and its receptors in the AD forebrain, the hypothesis is that local delivery of NGF to the nbM could revive the cholinergic circuitry and thereby restore cognitive functions. Since NGF does not pass through the blood-brain barrier, approaches involving cerebral injections of genetically modified cells or viral vectors or implantation of encapsulated cells in the nbM in AD patients have been used. These attempts have been partially successful but also have limitations, which are presented and discussed here. In conclusion, these trials point to the importance of further development of NGF-related therapies in AD.


Subject(s)
Alzheimer Disease , Basal Nucleus of Meynert , Alzheimer Disease/therapy , Cell- and Tissue-Based Therapy , Cholinergic Neurons , Genetic Therapy , Humans , Nerve Growth Factor
11.
J Biol Chem ; 294(29): 11154-11165, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31167782

ABSTRACT

Homocysteine, a metabolite of the methionine cycle, is a known agonist of N-methyl-d-aspartate receptor (NMDAR), a glutamate receptor subtype and is involved in NMDAR-mediated neurotoxicity. Our previous findings have shown that homocysteine-induced, NMDAR-mediated neurotoxicity is facilitated by a sustained increase in phosphorylation and activation of extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK MAPK). In the current study, we investigated the role GluN1/GluN2A-containing functional NMDAR (GluN2A-NMDAR) and GluN1/GluN2B-containing functional NMDAR (GluN2B-NMDAR) in homocysteine-induced neurotoxicity. Our findings revealed that exposing primary cortical neuronal cultures to homocysteine leads to a sustained low-level increase in intracellular Ca2+ We also showed that pharmacological inhibition of GluN2A-NMDAR or genetic deletion of the GluN2A subunit attenuates homocysteine-induced increase in intracellular Ca2+ Our results further established the role of GluN2A-NMDAR in homocysteine-mediated sustained ERK MAPK phosphorylation and neuronal cell death. Of note, the preferential role of GluN2A-NMDAR in homocysteine-induced neurotoxicity was distinctly different from glutamate-NMDAR-induced excitotoxic cell death that involves overactivation of GluN2B-NMDAR and is independent of ERK MAPK activation. These findings indicate a critical role of GluN2A-NMDAR-mediated signaling in homocysteine-induced neurotoxicity.


Subject(s)
Calcium/metabolism , Cell Death/drug effects , Homocysteine/pharmacology , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Enzyme Activation , Female , Ion Transport , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Phosphorylation , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics
12.
Front Neurosci ; 13: 38, 2019.
Article in English | MEDLINE | ID: mdl-30804738

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with abnormal protein modification, inflammation and memory impairment. Aggregated amyloid beta (Aß) and phosphorylated tau proteins are medical diagnostic features. Loss of memory in AD has been associated with central cholinergic dysfunction in basal forebrain, from where the cholinergic circuitry projects to cerebral cortex and hippocampus. Various reports link AD progression with declining activity of cholinergic neurons in basal forebrain. The neurotrophic molecule, nerve growth factor (NGF), plays a major role in the maintenance of cholinergic neurons integrity and function, both during development and adulthood. Numerous studies have also shown that NGF contributes to the survival and regeneration of neurons during aging and in age-related diseases such as AD. Changes in neurotrophic signaling pathways are involved in the aging process and contribute to cholinergic and cognitive decline as observed in AD. Further, gradual dysregulation of neurotrophic factors like NGF and brain derived neurotrophic factor (BDNF) have been reported during AD development thus intensifying further research in targeting these factors as disease modifying therapies against AD. Today, there is no cure available for AD and the effects of the symptomatic treatment like cholinesterase inhibitors (ChEIs) and memantine are transient and moderate. Although many AD treatment studies are being carried out, there has not been any breakthrough and new therapies are thus highly needed. Long-term effective therapy for alleviating cognitive impairment is a major unmet need. Discussion and summarizing the new advancements of using NGF as a potential therapeutic implication in AD are important. In summary, the intent of this review is describing available experimental and clinical data related to AD therapy, priming to gain additional facts associated with the importance of NGF for AD treatment, and encapsulated cell biodelivery (ECB) as an efficient tool for NGF delivery.

13.
Mol Neurobiol ; 56(7): 4601-4619, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30361890

ABSTRACT

OMI/HTRA2 (high-temperature requirement serine protease A2) is a mitochondrial serine protease involved in several cellular processes, including autophagy, chaperone activity, and apoptosis. Few studies on the role of OMI/HTRA2 in Alzheimer's disease (AD) are available, but none on its relationship with the cholinergic system and neurotrophic factors as well as other AD-related proteins. In this study, immunohistochemical analyses revealed that AD patients had a higher cytosolic distribution of OMI/HTRA2 protein compared to controls. Quantitative analyses on brain extracts indicated a significant increase in the active form of OMI/HTRA2 in the AD brain. Activated OMI/HTRA2 protein positively correlated with stress-associated read-through acetylcholinesterase activity. In addition, α7 nicotinic acetylcholine receptor gene expression, a receptor also known to be localized on the outer membrane of mitochondria, showed a strong correlation with OMI/HTRA2 gene expression in three different brain regions. Interestingly, the activated OMI/HTRA2 levels also correlated with the activity of the acetylcholine-biosynthesizing enzyme, choline acetyltransferase (ChAT); with levels of the neurotrophic factors, NGF and BDNF; with levels of the soluble fragments of amyloid precursor protein (APP); and with gene expression of the microtubule-associated protein tau in the examined brain regions. Overall, the results demonstrate increased levels of the mitochondrial serine protease OMI/HTRA2, and a coherent pattern of association between the activated form of OMI/HTRA2 and several key proteins involved in AD pathology. In this paper, we propose a new hypothetical model to highlight the importance and needs of further investigation on the role of OMI/HTRA2 in the mitochondrial function and AD.


Subject(s)
Acetylcholine/metabolism , Alzheimer Disease/enzymology , Brain/enzymology , High-Temperature Requirement A Serine Peptidase 2/metabolism , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Aged , Aged, 80 and over , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Biomarkers/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Butyrylcholinesterase/metabolism , Female , Gene Expression Regulation , High-Temperature Requirement A Serine Peptidase 2/genetics , Humans , Male , Middle Aged , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , tau Proteins/genetics , tau Proteins/metabolism
14.
Exp Cell Res ; 371(1): 175-184, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30092220

ABSTRACT

Alzheimer's disease (AD) is characterized by early degeneration of cholinergic neurons and decreased levels of nerve growth factor (NGF). Thus, increasing the NGF levels by for instance encapsulated cell bio-delivery (ECB) is a potential treatment strategy. The results from our previous first-in-human studies on ECB of NGF to the basal forebrain cholinergic neurons were promising, but indicated some variability of long-term viability of the encapsulated cells and associated reduced NGF-release. Here we studied the effect of amyloid beta-peptides (Aß), interleukin 1-beta (IL-1ß), and CSF from AD, Lewy body dementia (LBD) or subjective cognitive impairment (SCI) patients on the NGF overproducing cell line NGC-0295. At physiological concentrations, neither Aß40 nor Aß42 had any major impact on cell viability or NGF-production. In contrast, IL-1ß dose-dependently affected NGF-production over time. Exposure of NGF-producing cells to CSF from AD patients showed significantly reduced NGF-release as compared to CSF from LBD or SCI patients. By mass spectrometry we found 3 proteins involved in inflammatory pathways to have an altered expression in AD CSF compared to LBD and SCI. Cell survival and NGF-release were not affected by Aß. NGF-release was affected by IL-1ß, suggesting that inflammation has a negative effect on ECB cells.


Subject(s)
Alzheimer Disease/genetics , Cerebrospinal Fluid/chemistry , Gene Expression Regulation/drug effects , Interleukin-1beta/pharmacology , Nerve Growth Factor/genetics , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Cell Line, Transformed , Cell Survival/drug effects , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lewy Body Disease/cerebrospinal fluid , Lewy Body Disease/genetics , Lewy Body Disease/pathology , Nerve Growth Factor/metabolism , Peptide Fragments/pharmacology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Staurosporine/pharmacology
15.
Sci Rep ; 7(1): 7140, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28769045

ABSTRACT

Arsenic is globally infamous for inducing immunosuppression associated with prevalence of opportunistic infection in exposed population, although the mechanism remains elusive. In this study, we investigate the effect of arsenic exposure on thymocyte lineage commitment and the involvement of regulatory T cells (Treg) in arsenic-induced immunosuppression. Male Balb/c mice were exposed to 0.038, 0.38 and 3.8 ppm sodium arsenite for 7, 15 and 30 days through oral gavage. Arsenic exposure promoted CD4 lineage commitment in a dose dependent manner supported by the expression of ThPOK in thymus. Arsenic also increased splenic CD4+ T cells and promoted their differentiation into Treg cells. In parallel, arsenic exposure induced immunosuppression characterized by low cytokine secretion from splenocytes and increased susceptibility to Mycobacterium fortuitum (M. fortuitum) infection. Therefore, we linked arsenic-induced rise in Treg cells with suppressed Th1 and Th2 related cytokines, which has been reversed by inhibition of Treg cells in-vivo using wortmannin. Other parameters like body weight, kidney and liver function, histoanatomy of thymus and spleen as well as thymocyte and splenocytes viability were unaltered by arsenic exposure. Taken together our findings indicated that environmentally relevant dose of arsenic enhanced differentiation of Treg cells which in turn induce immunosuppression in experimental animals.


Subject(s)
Arsenic/pharmacology , CD4 Antigens/metabolism , Cytokines/biosynthesis , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Thymocytes/drug effects , Thymocytes/metabolism , Animals , Biomarkers , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Lymphocyte Count , Male , Mice , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , Thymocytes/immunology , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Environ Toxicol ; 32(10): 2295-2304, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28707438

ABSTRACT

Declining rate of human fertility is a growing concern, where lifestyle and environmental factors play an important role. We recently demonstrated that tributyltin (TBT), an omnipresent endocrine disruptor, affects testicular cells in vitro. In this study, male Wistar rats were gavaged a single dose of 10, 20, and 30 mg/kg TBT-chloride (TBTC) (to mimic accidental exposure in vivo) and sacrificed on day 3 and day 7, respectively. TBT bioavailability was evaluated by estimating total tin content, and essential metal levels were analyzed along with redox molecules (ROS and GSH/GSSG) to understand the effect on physiological conditions. Blood-testicular barrier (BTB) disruption, levels of associated proteins and activity of proteolytic enzymes were evaluated to understand the effect on BTB. Histological analysis of tissue architecture and effect on protein expression of steroidogenic, stress and apoptotic markers were also evaluated. Widespread TBTC pollution can be an eventual threat to male fertility worldwide.


Subject(s)
Endocrine Disruptors/toxicity , Fertility/drug effects , Trialkyltin Compounds/toxicity , Animals , Apoptosis , Body Weight/drug effects , Lipid Metabolism/drug effects , Male , Organ Size/drug effects , Oxidation-Reduction , Oxidative Stress/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism , Testis/drug effects , Testis/metabolism
17.
Drug Chem Toxicol ; 39(1): 74-80, 2016.
Article in English | MEDLINE | ID: mdl-25885549

ABSTRACT

CONTEXT: Cadmium (Cd) is known to cause severe damage to various organs including lung, liver, kidney, brain and reproductive system. Several studies have reported the induction of oxidative stress pathways following Cd exposure. OBJECTIVE: Since oxidative stress is also deemed responsible for inducing male infertility, a growing worldwide concern, we tried to understand whether the antioxidant N-acetylcysteine (NAC) can be a potential therapeutic agent to counter Cd toxicity using primary Leydig cells. MATERIALS AND METHODS: This study highlights the initial cellular alterations which culminate in cell death induction. Primary Leydig cells were isolated from 28-day-old male Wistar rats, exposed to various concentrations of Cd in vitro and biochemical and cell death parameters were evaluated to understand the effect of Cd. NAC pre-treatment was done to understand its protective efficacy. RESULTS: Following Cd exposure to Leydig cells in vitro, we found simultaneous intracellular calcium (Ca(2+)) increase and reduction in mitochondrial membrane polarization at 30 min, followed by significant induction of reactive oxygen species and MAPK-extracellular-regulated kinases with concurrent glutathione depletion at 1 h, and significant cell death (both necrotic and apoptotic) at 6 and 18 h, respectively. Pre-treatment with NAC abrogated all these toxic manifestations and showed significantly reduced cell death. NAC also rescued the expression of 3-ßHSD, a major steroidogenic protein. DISCUSSION AND CONCLUSION: Taken together, these data illustrated that NAC can be used as a potential protective agent against Cd-induced testicular toxicity, especially with regards to oxidative stress-induced Leydig cell toxicity.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Cadmium/toxicity , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Cadmium/administration & dosage , Calcium/metabolism , Cell Death/drug effects , Glutathione/metabolism , Leydig Cells/drug effects , Male , Mitochondrial Membranes/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Time Factors
18.
Chem Biol Interact ; 238: 138-50, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26079211

ABSTRACT

Spirulina is a widely used health supplement and is a dietary source of C-Phycocyanin (CPC), a potent anti-oxidant. We have previously reported the neurotoxic potential of tributyltin chloride (TBTC), an environmental pollutant and potent biocide. In this study, we have evaluated the protective efficacy of CPC against TBTC induced neurotoxicity. To evaluate the extent of neuroprotection offered by CPC, its efficacy was compared with the degree of protection offered by N-acetylcysteine (NAC) (a well known neuroprotective drug, taken as a positive control). Male Wistar rats (28 day old) were administered with 20mg/kg TBTC (oral) and 50mg/kg CPC or 50mg/kg NAC (i.p.), alone or in combination, and various parameters were evaluated. These include blood-brain barrier (BBB) damage; redox parameters (ROS, GSH, redox pathway associated enzymes, oxidative stress markers); inflammatory, cellular, and stress markers; apoptotic proteins and in situ cell death assay (TUNEL). We observed increased CPC availability in cortical tissue following its administration. Although BBB associated proteins like claudin-5, p-glycoprotein and ZO-1 were restored, CPC/NAC failed to protect against TBTC induced overall BBB permeability (Evans blue extravasation). Both CPC and NAC remarkably reduced oxidative stress and inflammation. NAC effectively modulated redox pathway associated enzymes whereas CPC countered ROS levels efficiently. Interestingly, CPC and NAC were equivalently capable of reducing apoptotic markers, astroglial activation and cell death. This study illustrates the various pathways involved in CPC mediated neuroprotection against this environmental neurotoxicant and highlights its capability to modulate glial cell activity.


Subject(s)
Acetylcysteine/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Neuroprotective Agents/pharmacology , Phycocyanin/pharmacology , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Glutathione/metabolism , Male , Oxidative Stress/drug effects , Oxidoreductases/metabolism , Permeability/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Trialkyltin Compounds/toxicity
19.
Environ Toxicol ; 30(12): 1393-405, 2015 Dec.
Article in English | MEDLINE | ID: mdl-24895210

ABSTRACT

Tributyltin (TBT), a well-known endocrine disruptor, is an omnipresent environmental pollutant and is explicitly used in many industrial applications. Previously we have shown its neurotoxic potential on cerebral cortex of male Wistar rats. As the effect of TBT on other brain regions is not known, we planned this study to evaluate its effect on four brain regions (cerebellum, hippocampus, hypothalamus, and striatum). Four-week-old male Wistar rats were gavaged with a single dose of TBT-chloride (TBTC) (10, 20, and 30 mg/kg) and sacrificed on days 3 and 7, respectively. Effect of TBTC on blood-brain barrier (BBB) permeability and tin (Sn) accumulation were measured. Oxidative stress indexes such as reactive oxygen species (ROS), reduced and oxidized glutathione (GSH/GSSG) ratio, lipid peroxidation, and protein carbonylation were analyzed as they play an imperative role in various neuropathological conditions. Since metal catalyzed reactions are a major source of oxidant generation, levels of essential metals like iron (Fe), zinc (Zn), and calcium (Ca) were estimated. We found that TBTC disrupted BBB and increased Sn accumulation, both of which appear significantly correlated. Altered metal homeostasis and ROS generation accompanied by elevated lipid peroxidation and protein carbonylation indicated oxidative damage which appeared more pronounced in the striatum than in cerebellum, hippocampus, and hypothalamus. This could be associated to the depleted GSH levels in striatum. These results suggest that striatum is more susceptible to TBTC induced oxidative damage as compared with other brain regions under study.


Subject(s)
Brain/drug effects , Trialkyltin Compounds/toxicity , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Calcium/analysis , Cerebral Cortex/drug effects , Corpus Striatum/metabolism , Glutathione/metabolism , Iron/analysis , Lipid Peroxidation/drug effects , Male , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Spectrophotometry, Atomic , Zinc/analysis
20.
Environ Toxicol Pharmacol ; 37(3): 1048-59, 2014 May.
Article in English | MEDLINE | ID: mdl-24762416

ABSTRACT

Tributyltin (TBT) is a potent biocide and commonly used in various industrial sectors. Humans are mainly exposed through the food chain. We have previously demonstrated tin accumulation in brain following TBT-chloride (TBTC) exposure. In this study, effect of TBTC on dissociated cells from different brain regions was evaluated. Cytotoxicity assay (MTT), mode of cell death (Annexin V/PI assay), oxidative stress parameters (ROS and lipid peroxidation), reducing power of the cell (GSH), mitochondrial membrane potential (MMP) and intracellular Ca(2+) were evaluated to ascertain the effect of TBTC. Expression of glial fibrillary acidic protein (GFAP) was measured to understand the effect on astroglial cells. TBTC as low as 30 nM was found to reduce GSH levels, whereas higher doses of 300 and 3000 nM induced ROS generation and marked loss in cell viability mainly through apoptosis. Striatum showed higher susceptibility than other regions, which may have further implications on various neurological aspects.


Subject(s)
Brain/cytology , Brain/drug effects , Disinfectants/toxicity , Trialkyltin Compounds/toxicity , Animals , Brain/metabolism , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...