Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 44(4): 477-490, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37988123

ABSTRACT

Neuronal loss in the ipsilateral thalamus after focal cortical infarction participates in post-stroke cognitive deficits, and enhanced angiogenesis in the thalamus is expected to reduce neuronal damage. We hypothesize that novel translocator protein (TSPO) ligand, 2-Cl-MGV-1, can promote angiogenesis, attenuate neuronal loss in the thalamus, and ameliorate post-stroke cognitive deficits. Cortical infarction was induced by distal middle cerebral artery occlusion (dMCAO) in stroke-prone renovascular hypertensive rats. 2-Cl-MGV-1 or dimethyl sulfoxide was administered 24 h after dMCAO and then for 6 or 13 days. Spatial learning and memory were assessed using the Morris water maze. Neuronal loss, TSPO expression, angiogenesis, and intrinsic pathway were determined by immunofluorescence and immunoblotting 7 and 14 days after dMCAO. Cortical infarction caused post-stroke cognitive deficits and secondary neuronal loss with gliosis in the ipsilateral thalamus within 14 days of dMCAO. Increased angiogenesis and elevated expression of vascular TSPO were detected in the ipsilateral thalamus, and treatment with 2-Cl-MGV-1 enhanced angiogenesis by stimulating the PI3K-AKT-mTOR pathway. The effects of 2-Cl-MGV-1 on angiogenesis coincided with reduced neuronal loss in the thalamus and contributed to improvements in post-stroke cognitive deficits. Our findings suggest that 2-Cl-MGV-1 stimulates angiogenesis, ameliorates neuronal loss in the thalamus, and improves post-stroke cognitive deficits.


Subject(s)
Angiogenesis , Carbamates , Quinazolines , Stroke , Rats , Animals , Rats, Sprague-Dawley , Ligands , Phosphatidylinositol 3-Kinases/metabolism , Infarction, Middle Cerebral Artery/complications , Stroke/metabolism , Thalamus/metabolism , Cognition
2.
Int J Mol Sci ; 21(20)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066585

ABSTRACT

Recent studies have shown that the selective estrogen receptor modulator (SERM) raloxifene had pronounced protective effects against progressing brain damage after traumatic brain injury (TBI) in mice. These studies, indicating beneficial effects of raloxifene for brain health, prompted the study of the history and present state of knowledge of this topic. It appears that, apart from raloxifene, to date, four nonrelated compounds have shown comparable beneficial effects-fucoidan, pifithrin, SMM-189 (5-dihydroxy-phenyl]-phenyl-methanone), and translocator protein (TSPO) ligands. Raloxifene, however, is ahead of the field, as for more than two decades it has been used in medical practice for various chronic ailments in humans. Thus, apart from different types of animal and cell culture studies, it has also been assessed in various human clinical trials, including assaying its effects on mild cognitive impairments. Regarding cell types, raloxifene protects neurons from cell death, prevents glial activation, ameliorates myelin damage, and maintains health of endothelial cells. At whole central nervous system (CNS) levels, raloxifene ameliorated mild cognitive impairments, as seen in clinical trials, and showed beneficial effects in animal models of Parkinson's disease. Moreover, with stroke and TBI in animal models, raloxifene showed curative effects. Furthermore, raloxifene showed healing effects regarding multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS) in cell culture. The adverse biological signals typical of these conditions relate to neuronal activity, neurotransmitters and their receptors, plasticity, inflammation, oxidative stress, nitric oxide, calcium homeostasis, cell death, behavioral impairments, etc. Raloxifene favorably modulates these signals toward cell health-on the one hand, by modulating gene expression of the relevant proteins, for example by way of its binding to the cell nuclear estrogen receptors ERα and ERß (genomic effects) and, on the other hand (nongenomic effects) by modulation of mitochondrial activity, reduction of oxidative stress and programmed cell death, maintaining metabolic balance, degradation of Abeta, and modulation of intracellular cholesterol levels. More specifically regarding Alzheimer's disease, raloxifene may not cure diagnosed Alzheimer's disease. However, the onset of Alzheimer's disease may be delayed or arrested by raloxifene's capability to attenuate mild cognitive impairment. Mild cognitive impairment is a condition that may precede diagnosis of Alzheimer's disease. In this review, relatively new insights are addressed regarding the notion that Alzheimer's disease can be caused by bacterial (as well as viral) infections, together with the most recent findings that raloxifene can counteract infections of at least some bacterial and viral strains. Thus, here, an overview of potential treatments of neurodegenerative disease by raloxifene is presented, and attention is paid to subcellular molecular biological pathways that may be involved.


Subject(s)
Brain Injuries/drug therapy , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Animals , Brain Injuries/metabolism , Humans , Neurodegenerative Diseases/metabolism
3.
Mar Drugs ; 18(5)2020 May 05.
Article in English | MEDLINE | ID: mdl-32380741

ABSTRACT

Increased interest in natural antioxidants has brought to light the fucoidans (sulfated polysaccharides present in brown marine algae) as highly valued nutrients as well as effective and safe therapeutics against several diseases. Based on their satisfactory in vitro antioxidant potency, researchers have identified this molecule as an efficient remedy for neuropathological as well as metabolic disorders. Some of this therapeutic activity is accomplished by upregulation of cytoprotective molecular pathways capable of restoring the enzymatic antioxidant activity and normal mitochondrial functions. Sirtuin-3 has been discovered as a key player for achieving the neuroprotective role of fucoidan by managing these pathways, whose ultimate goal is retrieving the entirety of the antioxidant response and preventing apoptosis of neurons, thereby averting neurodegeneration and brain injuries. Another pathway whereby fucoidan exerts neuroprotective capabilities is by interactions with P-selectin on endothelial cells, thereby preventing macrophages from entering the brain proper. Furthermore, beneficial influences of fucoidan have been established in hepatocytes after xenobiotic induced liver injury by decreasing transaminase leakage and autophagy as well as obtaining optimal levels of intracellular fiber, which ultimately prevents fibrosis. The hepatoprotective role of this marine polysaccharide also includes a sirtuin, namely sirtuin-1 overexpression, which alleviates obesity and insulin resistance through suppression of hyperglycemia, reducing inflammation and stimulation of enzymatic antioxidant response. While fucoidan is very effective in animal models for brain injury and neuronal degeneration, in general, it is accepted that fucoidan shows somewhat limited potency in liver. Thus far, it has been used in large doses for treatment of acute liver injuries. Thus, it appears that further optimization of fucoidan derivatives may establish enhanced versatility for treatments of various disorders, in addition to brain injury and disease.


Subject(s)
Antioxidants/pharmacology , Brain Injuries/drug therapy , Chemical and Drug Induced Liver Injury/drug therapy , Neuroprotective Agents/pharmacology , Polysaccharides/pharmacology , Sirtuins/metabolism , Animals , Antioxidants/therapeutic use , Brain/drug effects , Brain/pathology , Brain Injuries/pathology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Humans , Liver/drug effects , Liver/pathology , Neuroprotective Agents/therapeutic use , Phaeophyceae/chemistry , Polysaccharides/therapeutic use , Signal Transduction/drug effects
4.
Cells ; 9(4)2020 04 02.
Article in English | MEDLINE | ID: mdl-32252470

ABSTRACT

Neuroinflammation and cell death are among the common symptoms of many central nervous system diseases and injuries. Neuroinflammation and programmed cell death of the various cell types in the brain appear to be part of these disorders, and characteristic for each cell type, including neurons and glia cells. Concerning the effects of 18-kDa translocator protein (TSPO) on glial activation, as well as being associated with neuronal cell death, as a response mechanism to oxidative stress, the changes of its expression assayed with the aid of TSPO-specific positron emission tomography (PET) tracers' uptake could also offer evidence for following the pathogenesis of these disorders. This could potentially increase the number of diagnostic tests to accurately establish the stadium and development of the disease in question. Nonetheless, the differences in results regarding TSPO PET signals of first and second generations of tracers measured in patients with neurological disorders versus healthy controls indicate that we still have to understand more regarding TSPO characteristics. Expanding on investigations regarding the neuroprotective and healing effects of TSPO ligands could also contribute to a better understanding of the therapeutic potential of TSPO activity for brain damage due to brain injury and disease. Studies so far have directed attention to the effects on neurons and glia, and processes, such as death, inflammation, and regeneration. It is definitely worthwhile to drive such studies forward. From recent research it also appears that TSPO ligands, such as PK11195, Etifoxine, Emapunil, and 2-Cl-MGV-1, demonstrate the potential of targeting TSPO for treatments of brain diseases and disorders.


Subject(s)
Alcoholism/genetics , Brain Injuries, Traumatic/genetics , Brain/pathology , Mental Disorders/genetics , Neurodegenerative Diseases/genetics , Receptors, GABA/therapeutic use , Stroke/genetics , Humans
5.
Int J Mol Sci ; 20(3)2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30704062

ABSTRACT

The 18 kDa Translocator Protein (TSPO) is a marker for microglial activation as its expression is enhanced in activated microglia during neuroinflammation. TSPO ligands can attenuate neuroinflammation and neurotoxicity. In the present study, we examined the efficacy of new TSPO ligands designed by our laboratory, MGV-1 and 2-Cl-MGV-1, in mitigating an in vitro neuroinflammatory process compared to the classic TSPO ligand, PK 11195. We exposed BV-2 microglial cells to lipopolysaccharide (LPS) for 24 h to induce inflammatory response and added the three TSPO ligands: (1) one hour before LPS treatment (pretreatment), (2) simultaneously with LPS (cotreatment), and (3) one hour after LPS exposure (post-treatment). We evaluated the capability of TSPO ligands to reduce the levels of three glial inflammatory markers: cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and nitric oxide (NO). We compared the effects of the two novel ligands to PK 11195. Both 2-Cl-MGV-1 and MGV-1 reduced the levels of glial COX-2, iNOS, and NO in LPS-treated BV-2 cells more efficiently than PK 11195. Notably, even when added after exposure to LPS, all ligands were able to suppress the inflammatory response. Due to their pronounced anti-inflammatory activity, 2-Cl-MGV-1 and MGV-1 may serve as potential therapeutics in neuroinflammatory and neurodegenerative diseases.


Subject(s)
Carbamates/pharmacology , Inflammation/chemically induced , Inflammation/metabolism , Isoquinolines/pharmacology , Lipopolysaccharides/toxicity , Microglia/drug effects , Microglia/metabolism , Quinazolines/pharmacology , Receptors, GABA/metabolism , Animals , Blotting, Western , Cell Line , Mice
6.
Adv Pharmacol ; 82: 103-136, 2018.
Article in English | MEDLINE | ID: mdl-29413517

ABSTRACT

In 1999, the enigma of the 18kDa mitochondrial translocator protein (TSPO), also known as the peripheral-type benzodiazepine receptor, was the seeming disparity of the many functions attributed to TSPO, ranging from the potential of TSPO acting as a housekeeping gene at molecular biological levels to adaptations to stress, and even involvement in higher emotional and cognitive functioning, such as anxiety and depression. In the years since then, knowledge regarding the many functions modulated by TSPO has expanded, and understanding has deepened. In addition, new functions could be firmly associated with TSPO, such as regulation of programmed cell death and modulation of gene expression. Interestingly, control by the mitochondrial TSPO over both of these life and death functions appears to include Ca++ homeostasis, generation of reactive oxygen species (ROS), and ATP production. Other mitochondrial functions under TSPO control are considered to be steroidogenesis and tetrapyrrole metabolism. As TSPO effects on gene expression and on programmed cell death can be related to the wide range of functions that can be associated with TSPO, several of these five elements of Ca++, ROS, ATP, steroids, and tetrapyrroles may indeed form the basis of TSPO's capability to operate as a multifunctional housekeeping gene to maintain homeostasis of the cell and of the whole multicellular organism.


Subject(s)
Mitochondria/metabolism , Receptors, GABA/metabolism , Animals , Evolution, Molecular , Genes, Essential , Homeostasis , Humans , Reactive Oxygen Species/metabolism
7.
Exp Clin Endocrinol Diabetes ; 126(2): 77-84, 2018 02.
Article in English | MEDLINE | ID: mdl-28954307

ABSTRACT

The 18 kDa mitochondrial translocator protein (TSPO) ligands (10 µM), e. g., protoporphyrin IX, PK 11195 and FGIN-1-27, have different effects on metabolism and protein expression in human osteoblasts. In this study, we investigated the archetypical TSPO specific ligand Ro5-4864 (10 µM) effect in primary osteoblasts in culture aiming to further understand the TSPO role in these mature metabolically active cells.We found that following exposure to Ro5-4864, cellular [18F]-FDG incorporation and ATP content were reduced by 48% (p<0.001) and 44% (p<0.001), respectively. The mitochondrial membrane potential (ΔΨm) increased by 50% (p<0.01), mRNA synthesis of TSPO and voltage dependent anion channel (VDAC1) decreased both by 70%, the TSPO and VDAC1 protein expression decreased by 80% and 68%, respectively (p<0.001). Ro5 4864 caused a decrease in the proportion of cells in the G1 phase (by 20%, p<0.05), shifting the cell cycle to the S and G2/M phases. Furthermore, 63% decrease in hexokinase 2 protein expression (p<0.001) was found. However, we found no significant effects on hexokinase 2 mRNA expression (by RT-PCR). We also did not see significant changes in mitochondrial mass (MitoTracker Green assay), apoptosis rate (TUNEL assay), overall cell death (LDH assay), cellular proliferation (BrdU assay), cell maturation (cellular alkaline phosphatase assay), and the number of cells in the culture.Therefore, an overall effect of Ro5-4864 exhorts is via pathways related to the mitochondrial activity, which is only partly like PK 11195, but not to the other TSPO ligands.


Subject(s)
Benzodiazepinones/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Mitochondria/drug effects , Osteoblasts/drug effects , Receptors, GABA/drug effects , Cells, Cultured , Humans , In Vitro Techniques , Ligands
8.
Stroke ; 48(12): 3366-3374, 2017 12.
Article in English | MEDLINE | ID: mdl-29146879

ABSTRACT

BACKGROUND AND PURPOSE: Focal cortical infarction causes neuronal apoptosis in the ipsilateral nonischemic thalamus and hippocampus, which is potentially associated with poststroke cognitive deficits. TSPO (translocator protein) is critical in regulating mitochondrial apoptosis pathways. We examined the effects of the novel TSPO ligand 2-(2-chlorophenyl) quinazolin-4-yl dimethylcarbamate (2-Cl-MGV-1) on poststroke cognitive deficits, neuronal mitochondrial apoptosis, and secondary damage in the ipsilateral thalamus and hippocampus after cortical infarction. METHODS: One hundred fourteen hypertensive rats underwent successful distal middle cerebral artery occlusion (n=76) or sham procedures (n=38). 2-Cl-MGV-1 or dimethyl sulfoxide as vehicle was administrated 2 hours after distal middle cerebral artery occlusion and then for 6 or 13 days (n=19 per group). Spatial learning and memory were tested using the Morris water maze. Secondary degeneration and mitochondrial apoptosis in the thalamus and hippocampus were assessed using Nissl staining, immunohistochemistry, terminal deoxynucleotidyl transferase dUTP nick end labeling, JC-1 staining, and immunoblotting 7 and 14 days after surgery. RESULTS: Infarct volumes did not significantly differ between the vehicle and 2-Cl-MGV-1 groups. There were more neurons and fewer glia in the ipsilateral thalamus and hippocampus in the vehicle groups than in the sham-operated group 7 and 14 days post-distal middle cerebral artery occlusion. 2-Cl-MGV-1 significantly ameliorated spatial cognitive impairment and decreased neuronal death and glial activation when compared with vehicle treatment (P<0.05). The collapse of mitochondrial transmembrane potential and cytoplasmic release of apoptosis-inducing factors and cytochrome c was prevented within the thalamus. Caspase cleavage and the numbers of terminal deoxynucleotidyl transferase dUTP nick end labeling+ or Nissl atrophic cells were reduced within the thalamus and hippocampus. This was accompanied by upregulation of B-cell lymphoma 2 and downregulation of Bax (P<0.05). CONCLUSIONS: 2-Cl-MGV-1 reduces neuronal apoptosis via mitochondrial-dependent pathways and attenuates secondary damage in the nonischemic thalamus and hippocampus, potentially contributing to ameliorated cognitive deficits after cortical infarction.


Subject(s)
Apoptosis/drug effects , Carbamates/therapeutic use , Cerebral Infarction/drug therapy , Cerebral Infarction/psychology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/psychology , Hippocampus/pathology , Neuroprotective Agents/therapeutic use , Quinazolines/therapeutic use , Thalamus/pathology , Animals , Cerebral Infarction/pathology , Cognitive Dysfunction/etiology , Hippocampus/drug effects , Male , Maze Learning/drug effects , Membrane Potential, Mitochondrial/drug effects , Memory/drug effects , Mitochondria/drug effects , Neuroglia/drug effects , Neuroglia/pathology , Neurons/pathology , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Receptors, GABA/biosynthesis , Receptors, GABA/genetics , Thalamus/drug effects
9.
Int J Mol Sci ; 18(4)2017 Apr 07.
Article in English | MEDLINE | ID: mdl-28387723

ABSTRACT

It is known that knockdown of the mitochondrial 18 kDa translocator protein (TSPO) as well as TSPO ligands modulate various functions, including functions related to cancer. To study the ability of TSPO to regulate gene expression regarding such functions, we applied microarray analysis of gene expression to U118MG glioblastoma cells. Within 15 min, the classical TSPO ligand PK 11195 induced changes in expression of immediate early genes and transcription factors. These changes also included gene products that are part of the canonical pathway serving to modulate general gene expression. These changes are in accord with real-time, reverse transcriptase (RT) PCR. At the time points of 15, 30, 45, and 60 min, as well as 3 and 24 h of PK 11195 exposure, the functions associated with the changes in gene expression in these glioblastoma cells covered well known TSPO functions. These functions included cell viability, proliferation, differentiation, adhesion, migration, tumorigenesis, and angiogenesis. This was corroborated microscopically for cell migration, cell accumulation, adhesion, and neuronal differentiation. Changes in gene expression at 24 h of PK 11195 exposure were related to downregulation of tumorigenesis and upregulation of programmed cell death. In the vehicle treated as well as PK 11195 exposed cell cultures, our triple labeling showed intense TSPO labeling in the mitochondria but no TSPO signal in the cell nuclei. Thus, mitochondrial TSPO appears to be part of the mitochondria-to-nucleus signaling pathway for modulation of nuclear gene expression. The novel TSPO ligand 2-Cl-MGV-1 appeared to be very specific regarding modulation of gene expression of immediate early genes and transcription factors.


Subject(s)
Cell Nucleus/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Isoquinolines/pharmacology , Oligonucleotide Array Sequence Analysis/methods , Receptors, GABA/genetics , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Ligands , Mitochondria/genetics , Signal Transduction/drug effects
10.
Int J Mol Sci ; 17(6)2016 Jun 04.
Article in English | MEDLINE | ID: mdl-27271616

ABSTRACT

The 18 kDa translocator protein (TSPO) is highly 0conserved in eukaryotes and prokaryotes. Since its discovery in 1977, numerous studies established the TSPO's importance for life essential functions. For these studies, synthetic TSPO ligands typically are applied. Tetrapyrroles present endogenous ligands for the TSPO. Tetrapyrroles are also evolutionarily conserved and regulate multiple functions. TSPO and tetrapyrroles regulate each other. In animals TSPO-tetrapyrrole interactions range from effects on embryonic development to metabolism, programmed cell death, response to stress, injury and disease, and even to life span extension. In animals TSPOs are primarily located in mitochondria. In plants TSPOs are also present in plastids, the nuclear fraction, the endoplasmic reticulum, and Golgi stacks. This may contribute to translocation of tetrapyrrole intermediates across organelles' membranes. As in animals, plant TSPO binds heme and protoporphyrin IX. TSPO-tetrapyrrole interactions in plants appear to relate to development as well as stress conditions, including salt tolerance, abscisic acid-induced stress, reactive oxygen species homeostasis, and finally cell death regulation. In bacteria, TSPO is important for switching from aerobic to anaerobic metabolism, including the regulation of photosynthesis. As in mitochondria, in bacteria TSPO is located in the outer membrane. TSPO-tetrapyrrole interactions may be part of the establishment of the bacterial-eukaryote relationships, i.e., mitochondrial-eukaryote and plastid-plant endosymbiotic relationships.


Subject(s)
Eukaryota/metabolism , Prokaryotic Cells/metabolism , Receptors, GABA/metabolism , Tetrapyrroles/metabolism , Animals , Binding Sites , Biological Evolution , Biological Transport , Brain Diseases/metabolism , Humans , Insecta/metabolism , Ligands , Plants/metabolism , Protein Binding , Protoporphyrins/chemistry , Protoporphyrins/metabolism , Receptors, GABA/chemistry , Receptors, GABA/genetics , Structure-Activity Relationship , Tetrapyrroles/chemistry
11.
Cancer Genomics Proteomics ; 12(2): 77-81, 2015.
Article in English | MEDLINE | ID: mdl-25770191

ABSTRACT

BACKGROUND/AIM: Cigarette smoke (CS) is the main inducer of oral cancer, increasing the prevalence by 4-7 times. We examined induction of apoptosis by CS exposure of SCC-25 and SCC-15 oral cancer cells. MATERIALS AND METHODS: After controlled exposure to CS of various durations and at different time points we measured DNA fragmentation to assay apoptotic levels. RESULTS: SCC-15 cells showed a 70% (p<0.05) increase in apoptotic levels immediately after 30 min of exposure to CS. Twenty-four hours after 30-min CS exposure a further increase in apoptotic levels to 178% (p<0.05) could be observed. However, SCC-15 cells showed a decrease in apoptotic levels immediately after 180-min exposure to CS. CS-exposed SCC-25 cells did not show such CS-related effects. CONCLUSION: SCC-15 and SCC-25 oral cancer cells respond differently to CS regarding apoptotic cell death levels. In this respect, SCC-15 cells are sensitive to CS, while SCC-25 cells are not. Further comparisons between these cells may give insight regarding relationships between CS, apoptosis and invasiveness of oral cancer.


Subject(s)
DNA Fragmentation , Mouth Neoplasms/pathology , Neoplasms, Squamous Cell/pathology , Smoking/adverse effects , Apoptosis , Cell Line, Tumor , Humans
12.
Anticancer Agents Med Chem ; 14(4): 559-77, 2014 May.
Article in English | MEDLINE | ID: mdl-24628235

ABSTRACT

Many types of cancer, for example glioblastoma, show resistance against current anti-cancer treatments. One reason is that they are not capable to effectively activate their intracellular cell death pathways. Novel treatments designed to overcome these deficiencies in cancer cells present promising concepts to eradicate chemotherapy-resistant cancer cells. One of these approaches includes the membrane seeking compound erucylphosphohomocholine (ErPC3) which is part of the latest generation of alkylphospholipid analogs developed over the last two-and-a-half decades. ErPC3 exerts potent antineoplastic effects in animal models and against established cancer cell lines including, for example, glioblastoma and different types of leukemia, while sparing their normal counterparts. Starting with a historical survey, we report here on the anticancer activity of ErPC3 and on ErPC3's established mechanisms of action. We cover the current knowledge on the induction of mitochondrial apoptosis by ErPC3, including its interaction with the 18 kDa translocator protein (TSPO). In addition we discuss other signaling pathways modulated by ErPC3. Interaction with the TSPO leads to activation of the mitochondrial apoptosis cascade. This includes cardiolipin oxidation at mitochondrial levels, collapse of the mitochondrial membrane potential, and release of cytochrome c, the initiating steps of the mitochondrial apoptosis cascade. Other pathways modulated by ErPC3 include different kinases for the PI3K/Akt/mTOR and the MAP kinase pathways. Furthermore, ErPC3's cytotoxic actions may include its effects on phosphatidylcholine synthesis to inhibit the endoplasmic reticulum enzyme CTP:phosphocholine cytidyltransferase. These basic research data hopefully will lead to effective approaches toward exploitation of ErPC3 for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Erucic Acids/pharmacology , Mitochondrial Proteins/metabolism , Neoplasms/drug therapy , Phosphorylcholine/analogs & derivatives , Receptors, GABA/metabolism , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Membrane/metabolism , Erucic Acids/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Signal Transduction
13.
J Bioenerg Biomembr ; 46(3): 197-204, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532136

ABSTRACT

Ligands of 18 kDa mitochondrial translocator protein (TSPO) differ in their cellular effects. We hypothesize that different TSPO ligands might exert different cellular responses. Therefore, following previous studies that showed different cellular responses to two specific TSPO ligands, PK 11195 and protoporphyrin IX, in human osteoblast-like cells in vitro, we now report the cellular response to another specific TSPO ligand, FGIN-1-27 (10(-5) M) (MW 436 kDa), in order to characterize the effects of each TSPO ligand. We found in primary culture of the human osteoblast-like cells that cell numbers were decreased by an average of 30% (p < 0.001) following exposure to 10(-5) M of FGIN-1-27 in comparison to vehicle controls. Cellular [(18)F]-FDG incorporation and ATP content were suppressed, by an average of 43% (p < 0.001) and 83% (p < 0.001), respectively. Mitochondrial mass and ΔΨm increased by an average of 26% (p < 0.01) and 425% (p < 0.0001) respectively. Lactate dehydrogenase activity was enhanced in culture media by 60% (p < 0.05), indicating overall cell death, while no increase in apoptotic levels was observed. Cellular proliferation, as determined by BrdU assay, was not affected. Synthesis of mRNA of TSPO, VDAC 1, and hexokinase 2 decreased in 0.3, 0.3 and 0.5 fold respectively, with accompanying decreases in protein expression of TSPO and Voltage Dependent Anion Channel 1 by 23% (p < 0.001) and 98% (p < 0.001), respectively, but without changes in hexokinase 2 protein expression. Thus it appears that 10(-5) M FGIN-1-27 reduces cell viability, cell metabolism, and mitochondrial function. Previously we found similar effects of PK 11195 on mitochondrial function and cell metabolism and of protoporphyrin IX on cell death in primary osteoblast-like cells.


Subject(s)
Indoleacetic Acids/administration & dosage , Mitochondria/physiology , Osteoblasts/cytology , Osteoblasts/physiology , Receptors, GABA/drug effects , Receptors, GABA/metabolism , Aged , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured/drug effects , Cells, Cultured/physiology , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Mitochondria/drug effects , Osteoblasts/drug effects
14.
CNS Neurol Disord Drug Targets ; 13(4): 574-92, 2014.
Article in English | MEDLINE | ID: mdl-24168369

ABSTRACT

The role of the 18-kDa Translocator Protein (TSPO) in cell death induced by NH4Cl (1-50 mM) for 24-72 hours to human glioblastoma U118MG cells was investigated. Cell death was already observed after 48 hours of treatment with NH4Cl at 5 mM. Dose and time-responses curves indicated that 15 mM of NH4Cl applied for 72 hours was the optimal condition for our viability assays. For example, 72 hours of 15 mM of NH4Cl caused a 50.3% increase in propidium iodide uptake, and lactate dehydrogenase release was 41.2% of the positive control, indicating significant increases in cell death. Furthermore, compared to vehicle control, these experimental conditions resulted in a significant decrease of 44.9% of the mitochondrial activity, a 62.3% increase in incidence of collapse of mitochondrial membrane potential, and an increase of 49.0% of cardiolipin peroxidation. In addition, a significant 4.3 fold increase in the maximal binding capacity (Bmax) of TSPO was found in NH4Cl-exposed cells. Surprisingly, western blot analysis and real-time PCR did not demonstrate changes in TSPO expression. We also found that neither NH4Cl nor glutamine (a metabolic product of enhanced NH4Cl levels) inhibited binding of the TSPO ligand [(3)H]PK 11195. Interestingly, we observed a bimodal effect of the TSPO ligands PK 11195, Ro5-4864, and FGIN-1-27 on the toxicity of NH4Cl; such that 1-100 nM concentrations of TSPO ligands were protective, while concentrations above 1 µM enhanced NH4Cl-induced cell death processes. In conclusion, TSPO takes part in a bimodal way in the lethal effects induced by NH4Cl in glial type cells.


Subject(s)
Ammonium Chloride/toxicity , Cell Death/drug effects , Cell Death/physiology , Hyperammonemia/physiopathology , Receptors, GABA/metabolism , Benzodiazepinones/pharmacology , Cardiolipins/metabolism , Cell Line, Tumor , Cell Shape , Dose-Response Relationship, Drug , GABA Agents/pharmacology , Glutamine/metabolism , Humans , Hyperammonemia/chemically induced , Hyperammonemia/drug therapy , Indoleacetic Acids/pharmacology , Isoquinolines/metabolism , Isoquinolines/pharmacology , L-Lactate Dehydrogenase/metabolism , Ligands , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/drug effects , Mitochondria/physiology , Neuroprotective Agents/pharmacology , Propidium/chemistry
15.
J Bioenerg Biomembr ; 45(4): 333-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23475134

ABSTRACT

In several pathological conditions, when conversion of Protoporphyrin (PP)IX into heme is impaired, a toxic accumulation of PPIX might occur. PPIX has been found to have affinity to the mitochondrial Translocator Protein 18 kDa. Since it is known that TSPO is abundant in human osteoblast cells, thus we assumed that PPIX can affect cellular functions via interactions with TSPO in these cells. Therefore we aimed to study the metabolic responses of human osteoblast to a high (10⁻5 M) concentration of PPIX in vitro. We found that in primary culture of human osteoblast-like cells cell numbers decreased following exposure to PPIX(10⁻5 M). Cellular [¹8F]-FDG incorporation, mitochondrial mass, ATP content were suppressed, and ΔΨm collapsed. Lactate dehydrogenase activity was enhanced in culture media, indicating overall cell death, while no increase in apoptotic levels was observed. Cellular proliferation was not affected. Protein expression of TSPO, VDAC 1, and hexokinase 2 decreased, although the synthesis of mRNA for hexokinase 2 increased. Thus, PPIX(10⁻5 M) has a cytotoxic effect on human osteoblast-like cell in vitro. Since these cells remain viable following exposure to another TSPO ligand, PK 11195 (10⁻5 M), as observed previously by us, the mode of action of PPIX on osteoblast-like cells is not identical to that of PK 11195. Accordingly pathological accumulation of PPIX may cause necrosis of osteoblasts leading to bone mass loss. We show that this phenomenon is unrelated to iron overload.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Protoporphyrins/metabolism , Protoporphyrins/pharmacology , Adenosine Triphosphate/metabolism , Aged , Aged, 80 and over , Apoptosis/drug effects , Cell Culture Techniques , Cell Death/drug effects , Cell Proliferation/drug effects , Female , Glucose/metabolism , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Phosphorylation , Receptors, GABA/metabolism , Voltage-Dependent Anion Channel 1/metabolism
16.
Article in English | MEDLINE | ID: mdl-23368281

ABSTRACT

The 18 kDa translocator protein (TSPO) is able to modulate several mitochondria-related cell death processes due to close association with proteins and other molecules involved to the mitochondrial permeability transition pore. In this way, herein we review cell death mechanisms targeting mitochondria, including programmed cell death type I, II and III. Several proteins involved in these cell death processes and with a possible interplay with the TSPO are also discussed including the voltage dependent anion channel, the adenine nucleotide transporter, cardiolipin, and the Bcl-2 family proteins. Noteworthy, TSPO has been also implicated in various other functions including mitochondrial respiration, immune and phagocytic host-defense response, microglial activation, inflammation, cell growth and differentiation, cancer, cell proliferation, ischemia, and mental and neuropathological disorders. We focused in recent studies of the TSPO particularly on cancer and neurodegeneration, thus presenting the TSPO as a core element in the role of mitochondria in diseases and related processes. Clinical benefit may be attainable by increasing pharmacological knowledge related to the TSPO. Recent patents typically relate to diagnosis and treatment of TSPO-related pathological conditions including cancer, and inflammatory conditions, as well as disorders associated with central nervous system, such as neurodegeneration, convulsions, anxiety, mental disorders, and dementia.


Subject(s)
Mitochondria/metabolism , Receptors, GABA/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Death , Central Nervous System Agents/pharmacology , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Drug Design , Humans , Ligands , Mitochondria/drug effects , Mitochondria/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Patents as Topic , Receptors, GABA/drug effects , Signal Transduction
17.
Pharmacogenet Genomics ; 22(8): 606-19, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22732722

ABSTRACT

OBJECTIVE: Previously, several important roles for glutamate have been described for the biology of primary brain tumors. For example, glutamate has been suggested to promote glioma cell proliferation by the activation of the 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid (AMPA) subtype of glutamate receptors. In the present study, we determined the potential regulatory roles of the 18-kDa translocator protein (TSPO) in the glutamatergic system in relation to cell death of brain tumor cells through knockdown of the TSPO by genetic manipulation. MATERIALS AND METHODS: With microarray analysis and validation of gene expression of particular genes using real-time PCR, we found effects because of small inhibitory RNA knockdown of the TSPO in human U118MG glioblastoma cells on gene expression of glutamate receptors, glutamate transporters, and enzymes for glutamate metabolism. We also applied antisense RNA to silence TSPO in rat C6 glioblastoma cells and assayed the effects on DNA fragmentation, indicative of apoptosis, because of glutamate exposure. RESULTS: In particular, the effects of TSPO silencing in human U118MG cells related to glutamate metabolism indicate a net effect of a reduction in glutamate levels, which may potentially protect the cells in question from cell death. The TSPO knockdown in C6 cells showed that TSPO is required for the induction of apoptosis because of glutamate exposure. CONCLUSION: These findings show that interactions between the TSPO and the glutamatergic system may play a role in tumor development of glioblastoma cells. This may also have implications for our understanding of the involvement of the TSPO in secondary brain damage and neurodegenerative diseases.


Subject(s)
Cell Survival , Glutamic Acid , Receptors, GABA , Receptors, Glutamate , Animals , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Glioblastoma , Glutamic Acid/genetics , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Humans , Oligonucleotide Array Sequence Analysis/methods , RNA, Small Interfering , Rats , Receptors, GABA/genetics , Receptors, GABA/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism
18.
Pharmacogenet Genomics ; 22(7): 538-50, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22547081

ABSTRACT

BACKGROUND: It is known that the mitochondrial 18 kDa translocator protein (TSPO) is present in almost all peripheral tissues and also in glial cells in the brain. TSPO levels are typically enhanced in correlation with tumorigenesis of cancer cells including glioblastoma. Relevant for angiogenesis, TSPO is also present in almost all cells of the cardiovascular system. METHODS: We studied the effect of TSPO knockdown by siRNA on various aspects of tumor growth of U118MG glioblastoma cells in two in-vivo models: a nude mouse model with intracerebral implants of U118MG glioblastoma cells and implantation of U118MG glioblastoma cells on the chorionallantoic membrane (CAM) of chicken embryos. In vitro, we further assayed the influence of TSPO on the invasive potential of U118MG cells. RESULTS: TSPO knockdown increased tumor growth in both in-vivo models compared with the scrambled siRNA control. Angiogenesis was also increased by TSPO knockdown as determined by a CAM assay. TSPO knockdown led to a decrease in adhesion to the proteins of the extracellular matrix, including fibronectin, collagen I, collagen IV, laminin I, and fibrinogen. TSPO knockdown also led to an enhancement in the migratory capability of U118MG cells, as determined in a modified Boyden chamber. Application of the TSPO ligand 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide (PK 11195) at a concentration of 25 µmol/l in the in-vitro models yielded results similar to those obtained on TSPO knockdown. We found no effects of PK 11195 on TSPO protein expression. Interestingly, at low nmol/l concentrations (around 1 nmol/l), PK 11195 enhanced adhesion to collagen I, suggesting a bimodal concentration effect of PK 11195. CONCLUSION: Intact TSPO appears to be able to counteract the invasive and angiogenic characteristics related to the aggressiveness of U118MG glioblastoma cells in vivo and in vitro.


Subject(s)
Cell Proliferation , Glioblastoma/metabolism , Glioblastoma/pathology , Receptors, GABA/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Chick Embryo , Glioblastoma/blood supply , Humans , Isoquinolines/pharmacology , Ligands , Male , Mice , Mice, Nude , Neovascularization, Pathologic , RNA, Small Interfering/metabolism
19.
Apoptosis ; 17(7): 647-65, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22544277

ABSTRACT

Various studies have shown that several lethal agents induce cell death via the mitochondrial 18 kDa Translocator Protein (TSPO). In this study we tested the possibility that nitric oxide (NO) is the signaling component inducing the TSPO to initiate cell death process. Cell viability assays included Trypan blue uptake, propidium iodide uptake, lactate dehydrogenase release, and DNA fragmentation. These assays showed that application of the specific TSPO ligand PK 11195 reduced these parameters for the lethal effects of the NO donor sodium nitroprusside (SNP) by 41, 27, 40, and 42 %, respectively. TSPO silencing by siRNA also reduced the measured lethal effects of SNP by 50 % for all of these four assays. With 2,3-bis[2-methoxy-4-nitro-5-sulphophenyl]-2H-tetrazolium-5-carboxyanilide (XTT) changes in metabolic activity were detected. PK 11195 and TSPO knockdown fully prevented the reductions in XTT signal otherwise induced by SNP. Collapse of the mitochondrial membrane potential was studied with the aid of JC-1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolylcarbocyanine chloride). PK 11195 and TSPO knockdown reduced, respectively by 36 and 100 %, the incidence of collapse of the mitochondrial membrane potential otherwise induced by SNP. 10-N-Nonyl-Acridine Orange (NAO) was used to detect mitochondrial reactive oxygen species generation due to SNP. PK 11195 and TSPO knockdown reduced this effect of SNP by 65 and 100 %, respectively. SNP did not affect TSPO protein expression and binding characteristics, and also did not cause TSPO S-nitrosylation. However, ß-actin and various other proteins (not further defined) were S-nitrosylated. In conclusion, TSPO is required for the lethal and metabolic effects of the NO donor SNP, but TSPO itself is not S-nitrosylated.


Subject(s)
Carrier Proteins/metabolism , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Animals , Blotting, Western , Cell Death/drug effects , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Isoquinolines/pharmacology , Ligands , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Weight , Nitrosation/drug effects , Protein Binding/drug effects , Rats , Voltage-Dependent Anion Channels/metabolism
20.
J Bioenerg Biomembr ; 43(6): 739-46, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22127435

ABSTRACT

The role of the TSPO in metabolism of human osteoblasts is unknown. We hypothesized that human osteoblast metabolism may be modulated by the TSPO. Therefore we evaluated the presence of TSPO in human osteoblast-like cells and the effect of its synthetic ligand PK 11195 on these cells. The presence of TSPO was determined by [(3)H]PK 11195 binding using Scatchard analysis: Bmax 7682 fmol/mg, Kd 9.24 nM. PK 11195 did not affect significantly cell proliferation, cell death, cellular viability, maturation, [(18)F]-FDG incorporation and hexokinase 2 gene expression or protein levels. PK 11195 exerted a suppressive effect on VDAC1 and caused an increase in TSPO gene expression or protein levels. In parallel there was an increase in mitochondrial mass, mitochondrial ATP content and a reduction in ΔΨm collapse. Thus, it appears that PK11195 (10(-5) M) stimulates mitochondrial activity in human osteoblast-like cells without affecting glycolytic activity and cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Isoquinolines/pharmacology , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Receptors, GABA/biosynthesis , Adenosine Triphosphate/metabolism , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Hexokinase/biosynthesis , Humans , Osteoblasts , Voltage-Dependent Anion Channel 1/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...