Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Support Care Cancer ; 29(1): 533-534, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32940769

ABSTRACT

This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1007/s00520-020-05770-w.

2.
Support Care Cancer ; 25(4): 1279-1289, 2017 04.
Article in English | MEDLINE | ID: mdl-27966023

ABSTRACT

PURPOSE: High-dose chemotherapy prior to autologous stem cell transplantation (ASCT) leads to adverse effects including mucositis, neutropenia and bacteremia. To reduce the toxicity, we treated myeloma and lymphoma patients with peroral bismuth as an adjuvant to chemotherapy to convey cytoprotection in non-malignant cells. METHODS: This trial was a prospective, randomised, double-blind, placebo-controlled pilot study of hematological inpatients (n = 50) receiving bismuth or placebo tablets, in order to identify any potential superiority of bismuth on toxicity from chemotherapy. RESULTS: We show for the first time that bismuth significantly reduces grade 2 stomatitis, febrile neutropenia and infections caused by melphalan in multiple myeloma, where adverse effects also were significantly linked to gender. In lymphoma patients, bismuth significantly reduces diarrhoea relative to placebo. Also, lymphoma patients' adverse effects were linked to gender. For the first time, bismuth is demonstrated as a safe strategy against chemotherapy's toxicity without interfering with intentional anti-cancer efficiency. Also, we show how gender significantly influences various adverse effects and response to treatment in both multiple myeloma and malignant lymphomas. CONCLUSION: These results may impact clinical prevention of chemotherapy's cytotoxicity in certain patient groups, and also, this study may direct further attention towards the impact of gender during the course and treatment outcome of malignant disorders.


Subject(s)
Bismuth/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Transplantation Conditioning/methods , Transplantation, Autologous/methods , Aged , Bismuth/administration & dosage , Bismuth/pharmacology , Double-Blind Method , Female , Humans , Lymphoma/pathology , Male , Middle Aged , Multiple Myeloma/pathology , Pilot Projects , Prospective Studies , Treatment Outcome
3.
Histochem Cell Biol ; 138(5): 787-802, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22820857

ABSTRACT

Multiple sclerosis (MS) is the most common neurodegenerative disease in the Western world affecting younger, otherwise healthy individuals. Today no curative treatment exists. Patients suffer from recurring attacks caused by demyelination and underlying neuroinflammation, ultimately leading to loss of neurons. Recent research shows that bio-liberation of gold ions from metallic gold implants can ameliorate inflammation, reduce apoptosis and promote proliferation of neuronal stem cells (NSCs) in a mouse model of focal brain injury. Based on these findings, the present study investigates whether metallic gold implants affect the clinical signs of disease progression and the pathological findings in experimental autoimmune encephalomyelitis (EAE), a rodent model of MS. Gold particles 20-45 µm suspended in hyaluronic acid were bilaterally injected into the lateral ventricles (LV) of young Lewis rats prior to EAE induction. Comparing gold-treated animals to untreated and vehicle-treated ones, a statistically significant slowing of disease progression in terms of reduced weight loss was seen. Despite massive inflammatory infiltration, terminal deoxynucleotidyl transferase dUTP nick end labeling staining revealed reduced apoptotic cell death in disease foci in the brain stem of gold-treated animals, alongside an up-regulation of glial fibrillary acidic protein-positive reactive astrocytes near the LV and in the brain stem. Cell counting of frizzled-9 and nestin-stained cells showed statistically significant up-regulation of NSCs migrating from the subventricular zone. Additionally, the neuroprotective proteins Metallothionein-1 and -2 were up-regulated in the corpus callosum. In conclusion, this study is the first to show that the presence of small gold implants affect disease progression in a rat model of MS, increasing the neurogenic response and reducing the loss of cells in disease foci. Gold implants might thus improve clinical outcome for MS patients and further research into the long-term effects of such localized gold treatment is warranted.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Gliosis/chemically induced , Gold/therapeutic use , Multiple Sclerosis/drug therapy , Animals , Brain Stem/chemistry , Brain Stem/drug effects , Brain Stem/pathology , Cell Movement/drug effects , Corpus Callosum/chemistry , Corpus Callosum/drug effects , Corpus Callosum/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glial Fibrillary Acidic Protein/analysis , Gliosis/pathology , Intermediate Filament Proteins/analysis , Lateral Ventricles/drug effects , Lateral Ventricles/pathology , Metallothionein/analysis , Multiple Sclerosis/pathology , Nerve Tissue Proteins/analysis , Nestin , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Rats , Rats, Inbred Lew , Receptors, Neurotransmitter/analysis , Up-Regulation/drug effects
4.
Basic Clin Pharmacol Toxicol ; 109(1): 1-10, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21205224

ABSTRACT

Silver is a metal with well-known antibacterial effects. This makes silver an attractive coating material for medical devices for use inside the body, e.g. orthopaedic prostheses and catheters used in neurosurgery as it has been found to reduce the high risk of infections. Lately, the use of nano-silver particles in the industry, e.g. woven into fabrics and furniture has increased, and thus the exposure to silver particles in daily life increases. To study the effect of metallic silver particles on nervous tissue, we injected micron-sized silver particles into the mouse brain by stereotactic procedures. After 7, 14 days and 9 months, the silver-exposed animals had considerable brain damage seen as cavity formation and inflammation adjacent to the injected metallic silver particles. The tissue loss involved both cortical and hippocampal structures and resulted in enlargement of the lateral ventricles. Autometallographic silver enhancement showed silver uptake in lysosomes of glia cells and neurons in the ipsilateral cortex and hippocampus alongside a minor uptake on the contralateral side. Silver was also detected in ependymal cells and the choroid plexus. After 9 months, spreading of silver to the kidneys was seen. Cell counts of immunostained sections showed that metallic silver induced a statistically significant inflammatory response, i.e. increased microgliosis (7 days: p < 0.0001; 14 days: p < 0.01; 9 months: p < 0.0001) and TNF-α expression (7 and 14 days: p < 0.0001; 9 months: p = 0.91). Significant astrogliosis (7, 14 days and 9 months: p < 0.0001) and increased metallothionein (MT I + II) expression (7 and 14 days: p < 0.0001; 9 months: p < 0.001) were also seen in silver-exposed brain tissue. We conclude that metallic silver implants release silver ions causing neuroinflammation and a progressive tissue loss in the brain.


Subject(s)
Brain/drug effects , Inflammation/chemically induced , Silver/toxicity , Animals , Brain/pathology , Cell Count , Female , Gene Expression Regulation/drug effects , Inflammation/pathology , Metallothionein/drug effects , Metallothionein/genetics , Metallothionein/metabolism , Mice , Mice, Inbred BALB C , Prostheses and Implants , Silver/pharmacokinetics , Stereotaxic Techniques , Time Factors , Tissue Distribution
5.
Exp Parasitol ; 127(2): 500-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21044627

ABSTRACT

Cerebral malaria (CM) causes substantial mortality and neurological sequelae in survivors, and no neuroprotective regimens are currently available for this condition. Erythropoietin (EPO) reduces neuropathology and improves survival in murine CM. Using the Plasmodium berghei model of CM, we investigated if EPO's neuroprotective effects include activation of endogenous neural stem cells (NSC). By using immunohistochemical markers of different NSC maturation stages, we show that EPO increased the number of nestin(+) cells in the dentate gyrus and in the sub-ventricular zone of the lateral ventricles, relative to control-treatment. 75% of the EPO-treated CM mice displayed migration as nestin(+) NSC. The NSC showed differentiation towards a neural cell lineage as shown by PSA-NCAM binding and NSC maturation and lineage commitment was significantly affected by exogenous EPO and by CM in the sub ventricular zone. These results indicate a rapid, EPO-dependent activation of NSC during CM pathology.


Subject(s)
Erythropoietin/therapeutic use , Malaria, Cerebral/drug therapy , Neural Stem Cells/drug effects , Neuroprotective Agents/therapeutic use , Plasmodium berghei , Analysis of Variance , Animals , Disease Models, Animal , Erythropoietin/pharmacology , Female , Immunohistochemistry , Intermediate Filament Proteins/analysis , Malaria, Cerebral/pathology , Malaria, Cerebral/physiopathology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/analysis , Nestin , Neural Cell Adhesion Molecule L1/analysis , Neural Stem Cells/chemistry , Neural Stem Cells/cytology , Neurites/physiology , Neuroprotective Agents/pharmacology , Sialic Acids/analysis , Specific Pathogen-Free Organisms
6.
Cerebrospinal Fluid Res ; 7: 20, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21054845

ABSTRACT

BACKGROUND: The water channel protein aquaporin-4 (AQP4) is reported to be of possible major importance for accessory cerebrospinal fluid (CSF) circulation pathways. We hypothesized that changes in AQP4 expression in specific brain regions correspond to the severity and duration of hydrocephalus. METHODS: Hydrocephalus was induced in adult rats (~8 weeks) by intracisternal kaolin injection and evaluated after two days, one week and two weeks. Using magnetic resonance imaging (MRI) we quantified lateral ventricular volume, water diffusion and blood-brain barrier properties in hydrocephalic and control animals. The brains were analysed for AQP4 density by western blotting and localisation by immunohistochemistry. Double fluorescence labelling was used to study cell specific origin of AQP4. RESULTS: Lateral ventricular volume was significantly increased over control at all time points after induction and the periventricular apparent diffusion coefficient (ADC) value significantly increased after one and two weeks of hydrocephalus. Relative AQP4 density was significantly decreased in both cortex and periventricular region after two days and normalized after one week. After two weeks, periventricular AQP4 expression was significantly increased. Relative periventricular AQP4 density was significantly correlated to lateral ventricular volume. AQP4 immunohistochemical analysis demonstrated the morphological expression pattern of AQP4 in hydrocephalus in astrocytes and ventricular ependyma. AQP4 co-localized with astrocytic glial fibrillary acidic protein (GFAP) in glia limitans. In vascular structures, AQP4 co-localized to astroglia but not to microglia or endothelial cells. CONCLUSIONS: AQP4 levels are significantly altered in a time and region dependent manner in kaolin-induced hydrocephalus. The presented data suggest that AQP4 could play an important neurodefensive role, and may be a promising future pharmaceutical target in hydrocephalus and CSF disorders.

7.
Glia ; 58(10): 1220-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20544857

ABSTRACT

Neuroglobin (Ngb) is proposed to be a neuron-specific, hypoxia-responsive, neuroprotective protein. However, results are conflicting concerning both Ngb's physiological and pathological significance. This study was designed to investigate the in vivo localization and regulation of Ngb in different neuropathological models representing traumatic injury, infectious, autoimmune, and excitotoxic pathogeneses. We profiled Ngb immunohistochemistry in murine models of traumatic brain injury, cerebral malaria, experimental autoimmune encephalitis, and kainic acid (KA)-mediated epileptic seizures that, to our knowledge, have not been studied in the context of Ngb. In control mice Ngb was expressed exclusively in neurons. In all pathological models except KA, in addition to neurons Ngb was present in reactive astrocytes. Ngb positive astrocytes were found within regions associated with most severe pathology and the astroglial scar. This is the first report of Ngb present in reactive astroglia and in scar-forming astrocytes in response to different pathological conditions relevant to human disease. In light of previously reported cyto-protective properties of Ngb, further insight may result in therapeutic ramifications.


Subject(s)
Astrocytes/metabolism , Autoimmune Diseases of the Nervous System/metabolism , Brain Injuries/metabolism , Encephalitis/metabolism , Globins/metabolism , Malaria, Cerebral/metabolism , Nerve Tissue Proteins/metabolism , Animals , Astrocytes/pathology , Autoimmune Diseases of the Nervous System/pathology , Brain/metabolism , Brain/pathology , Brain Injuries/pathology , Disease Models, Animal , Encephalitis/pathology , Epilepsy/chemically induced , Epilepsy/metabolism , Epilepsy/pathology , Female , Immunohistochemistry , Kainic Acid , Malaria, Cerebral/pathology , Male , Mice , Mice, Inbred C57BL , Neuroglobin , Neurons/metabolism , Neurons/pathology
8.
Ugeskr Laeger ; 172(17): 1293-6, 2010 Apr 26.
Article in Danish | MEDLINE | ID: mdl-20444397

ABSTRACT

Brain inflammatory responses exert dual roles leading to acute removal of debris and protracted (secondary) neurodegeneration and apoptosis. Proinflammatory mechanisms involve cytokines, oxidants, enzymes, and toxins that sustain pathology and that impede neural stem cell activation and regeneration. Nevertheless, the phase of secondary damage represents a window of opportunity for neuroprotective intervention. However, agents modulating a single target have failed. The multifaceted pathophysiology points towards a drug with pleiotropic, neuroprotective effects and/or abilities to activate neurogenesis.


Subject(s)
Brain Diseases , Inflammation , Nerve Degeneration , Nerve Regeneration , Blood-Brain Barrier/physiology , Brain Diseases/immunology , Brain Diseases/pathology , Brain Diseases/physiopathology , Encephalitis/immunology , Encephalitis/pathology , Encephalitis/physiopathology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/physiopathology , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Regeneration/immunology , Nerve Regeneration/physiology , Stem Cells/immunology , Stem Cells/physiology
9.
PLoS One ; 5(4): e10131, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20396380

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is one of the leading causes of disability and death among young people. Although much is already known about secondary brain damage the full range of brain tissue responses to TBI remains to be elucidated. A population of neurons located in cerebral areas associated with higher cognitive functions harbours a vesicular zinc pool co-localized with glutamate. This zinc enriched pool of synaptic vesicles has been hypothesized to take part in the injurious signalling cascade that follows pathological conditions such as seizures, ischemia and traumatic brain injury. Pathological release of excess zinc ions from pre-synaptic vesicles has been suggested to mediate cell damage/death to postsynaptic neurons. METHODOLOGY/PRINCIPAL FINDINGS: In order to substantiate the influence of vesicular zinc ions on TBI, we designed a study in which damage and zinc movements were analysed in several different ways. Twenty-four hours after TBI ZnT3-KO mice (mice without vesicular zinc) were compared to littermate Wild Type (WT) mice (mice with vesicular zinc) with regard to histopathology. Furthermore, in order to evaluate a possible neuro-protective dimension of chemical blocking of vesicular zinc, we treated lesioned mice with either DEDTC or selenite. Our study revealed that chemical blocking of vesicular zinc ions, either by chelation with DEDTC or accumulation in zinc-selenium nanocrystals, worsened the effects on the aftermath of TBI in the WT mice by increasing the number of necrotic and apoptotic cells within the first 24 hours after TBI, when compared to those of chemically untreated WT mice. CONCLUSION/SIGNIFICANCE: ZnT3-KO mice revealed more damage after TBI compared to WT controls. Following treatment with DEDTC or selenium an increase in the number of both dead and apoptotic cells were seen in the controls within the first 24 hours after TBI while the degree of damage in the ZnT3-KO mice remained largely unchanged. Further analyses revealed that the damage development in the two mouse strains was almost identical after either zinc chelation or zinc complexion therapy.


Subject(s)
Brain Injuries/metabolism , Central Nervous System/pathology , Neurons/pathology , Zinc/deficiency , Animals , Brain Injuries/pathology , Cell Death/drug effects , Central Nervous System/metabolism , Chelating Agents/pharmacology , Ditiocarb/analogs & derivatives , Ditiocarb/pharmacology , Glutamic Acid , Mice , Mice, Knockout , Neurons/drug effects , Sodium Selenite/pharmacology , Zinc/metabolism
10.
Invest Ophthalmol Vis Sci ; 51(9): 4809-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20357188

ABSTRACT

PURPOSE: To examine in rodent models of retinitis pigmentosa (RP) the expression of the neuroprotectants metallothionein-I and -II and of megalin, an endocytic receptor that mediates their transport into neurons. METHODS: Gene and protein expression were analyzed in retinas of rd1 and rds mice and in those of RCS (Royal College of Surgeons) rats of various ages. Glial cell markers (cellular retinaldehyde binding protein, CRALBP; glial fibrillary acidic protein, GFAP; CD11b; and isolectin B4) were used to establish the identity of the cells. RESULTS: Metallothionein-I and -II gene expression increased with age in normal and degenerating retinas and was significantly greater in the latter. Protein expression, corresponding to metallothionein-I+II, was first observed in rd1 mice in Müller cells at postnatal day (P)12 and in rds mice at P16, coinciding with the onset of GFAP expression in these cells. In RCS rats, the same distribution was observed, but not until P32, long after the onset of GFAP expression. Metallothionein-I+II was observed also in a small number of microglial cells. Megalin was expressed in the nerve fiber layer and in the region of the inner and outer segments in normal animals, but expression in the outer retina was lost with age in degenerating retinas. CONCLUSIONS: Induction of metallothionein-I and -II occurs in the RP models studied and correlates with glial activation. The progressive loss of megalin suggests that transport of metallothionein-I+II into the degenerating photoreceptors (from e.g., Müller cells), could be impaired, potentially limiting the actions of these metallothioneins.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Metallothionein/genetics , Retinitis Pigmentosa , Animals , Astrocytes/metabolism , Astrocytes/pathology , Avidin/metabolism , Biomarkers/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Disease Models, Animal , Glial Fibrillary Acidic Protein , In Situ Nick-End Labeling , Metallothionein/metabolism , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Microglia/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
11.
Brain Res ; 1307: 1-13, 2010 Jan 11.
Article in English | MEDLINE | ID: mdl-19840777

ABSTRACT

The discovery of neural stem cells (NSCs) provides new therapeutic strategies for brain injury by means of endogenous cell renewal. In the injured mouse brain, bio-liberated gold ions from gold implants mediate anti-inflammatory and antiapoptotic effects and activation of NSCs. This paper investigates the neuroprotective effects of gold following brain injury in mice. We show for the first time that endogenous NSCs express macrophage colony-stimulating factor (M-CSF) as part of their post-injury activation and that gold implants increase this response. Also, gold increases expression of neurotrophin (NT)-4, transforming growth factor-beta 3 (TGF-beta 3), leukemia inhibitory factor (LIF) and metallothionein I+II (MT-I+II) post-injury. This paper shows that gold ions modulate neurotrophic factors after injury and that hematopoietic factor M-CSF is expressed in activated NSCs.


Subject(s)
Adult Stem Cells/drug effects , Brain Injuries/metabolism , Gene Expression Regulation/drug effects , Gold/pharmacology , Macrophage Colony-Stimulating Factor/metabolism , Nerve Growth Factors/metabolism , Animals , Brain Injuries/pathology , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Lateral Ventricles/pathology , Macrophage Colony-Stimulating Factor/genetics , Metallothionein/metabolism , Mice , Mice, Inbred C57BL , Time Factors
12.
Leuk Lymphoma ; 51(2): 314-28, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20038220

ABSTRACT

Primary central nervous system lymphoma (PCNSL) in immunocompetent patients is highly malignant and has a poor prognosis. The PCNSL molecular features are reminiscent to some degree of diffuse large B-cell lymphoma (DLBCL), yet PCNSL shows unique molecular profiles and a distinct clinical behavior. This article characterizes the histopathology and expression profiles of metallothionein-I + II (MT-I + II) and their receptor megalin along with proliferation, oxidative stress, and apoptosis in PCNSL and in central nervous system (CNS) lymphomas due to relapse from DLBCL (collectively referred to as CNS lymphoma). We show for the first time that MT-I + II and megalin are significantly altered in CNS lymphoma relative to controls (reactive lymph nodes and non-lymphoma brain tissue with neuropathology). MT-I + II are secreted in the CNS and are found mainly in the lymphomatous cells, while their receptor megalin is increased in cerebral cells. This morphology likely reflects the CNS lymphoma microenvironment and molecular interactions between lymphomatous and neuronal cells.


Subject(s)
Central Nervous System Neoplasms/pathology , Low Density Lipoprotein Receptor-Related Protein-2/analysis , Lymphoma, Large B-Cell, Diffuse/pathology , Metallothionein/analysis , Oxidative Stress , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Cycle Proteins/analysis , Cell Proliferation/drug effects , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/metabolism , DNA-Binding Proteins/analysis , Guanine/analogs & derivatives , Guanine/analysis , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Malondialdehyde/analysis , Middle Aged , Minichromosome Maintenance Complex Component 7 , Nuclear Proteins/analysis , Tyrosine/analogs & derivatives , Tyrosine/analysis
13.
Biofactors ; 35(4): 315-25, 2009.
Article in English | MEDLINE | ID: mdl-19655389

ABSTRACT

Metallothionein (MT)-I+II synthesis is induced in the central nervous system (CNS) in response to practically any pathogen or disorder, where it is increased mainly in reactive glia. MT-I+II are involved in host defence reactions and neuroprotection during neuropathological conditions, in which MT-I+II decrease inflammation and secondary tissue damage (oxidative stress, neurodegeneration, and apoptosis) and promote post-injury repair and regeneration (angiogenesis, neurogenesis, neuronal sprouting and tissue remodelling). Intracellularly the molecular MT-I+II actions involve metal ion control and scavenging of reactive oxygen species (ROS) leading to cellular redox control. By regulating metal ions, MT-I+II can control metal-containing transcription factors, zinc-finger proteins and p53. However, the neuroprotective functions of MT-I+II also involve an extracellular component. MT-I+II protects the neurons by signal transduction through the low-density lipoprotein family of receptors on the cell surface involving lipoprotein receptor-1 (LRP1) and megalin (LRP2). In this review we discuss the newest data on cerebral MT-I+II functions following brain injury and experimental autoimmune encephalomyelitis.


Subject(s)
Metallothionein/physiology , Neuroprotective Agents , Animals , Brain/drug effects , Brain/metabolism , Brain Injuries/physiopathology , Encephalitis/prevention & control , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Erythropoietin/physiology , Humans , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Metallothionein/biosynthesis , Multiple Sclerosis/physiopathology , Neurodegenerative Diseases/drug therapy , Oxidative Stress/drug effects , Receptors, Cell Surface/physiology , Signal Transduction
14.
J Neurosci Res ; 87(13): 2926-36, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19405100

ABSTRACT

Metallothionein (MT) belongs to a family of metal-binding cysteine-rich proteins comprising several structurally related proteins implicated in tissue protection and regeneration after injuries and functioning as antiapoptotic antioxidants in neurological disorders. This has been demonstrated in animals receiving MT treatment and in mice with endogenous MT overexpression or null mutation during various experimental models of neuropathology, and also in patients with Alzheimer's disease and amyotrophic lateral sclerosis. Exogenously applied MT increases neurite outgrowth and neuronal survival in rat cerebellar, hippocampal, dopaminergic, and cortical neurons in vitro. In this study, the intraneuronal signaling involved in MT-mediated neuritogenesis was examined. The MT-induced neurite outgrowth in cultures of cerebellar granule neurons was dependent on activation of a heterotrimeric G-protein-coupled pathway but not on protein tyrosine kinases or on receptor tyrosine kinases. Activation of phospholipase C was necessary for MT-induced neurite outgrowth, and furthermore it was shown that inhibition of several intracellular protein kinases, such as protein kinase A, protein kinase C, phosphatidylinositol 3-kinase, Ca(2+)/calmodulin kinase-II, and mitogen-activated protein kinase kinase, abrogated the MT-mediated neuritogenic response. In addition, exogenously applied MT resulted in a decrease in phosphorylation of intraneuronal kinases implicated in proinflammatory reactions and apoptotic cell death, such as glycogen synthase-serine kinase 3alpha, Jun, and signal transducer and activator of transcription 3. This paper elucidates the intraneuronal molecular signaling involved in neuroprotective effects of MT.


Subject(s)
Metallothionein/pharmacology , Nerve Tissue Proteins/physiology , Neurites/drug effects , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured/drug effects , Cerebellum/cytology , Drug Evaluation, Preclinical , Heterotrimeric GTP-Binding Proteins/physiology , Metallothionein/administration & dosage , Metallothionein/physiology , Neuroprotective Agents/administration & dosage , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/physiology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-jun/physiology , Rabbits , Rats , Rats, Wistar , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/physiology
15.
Prog Histochem Cytochem ; 44(1): 1-27, 2009.
Article in English | MEDLINE | ID: mdl-19348909

ABSTRACT

In traumatic brain injury (TBI), the primary, irreversible damage associated with the moment of impact consists of cells dying from necrosis. This contributes to fuelling a chronic central nervous system (CNS) inflammation with increased formation of proinflammatory cytokines, enzymes and reactive oxygen species (ROS). ROS promote oxidative stress, which leads to neurodegeneration and ultimately results in programmed cell death (secondary injury). Since this delayed, secondary tissue loss occurs days to months following the primary injury it provides a therapeutic window where potential neuroprotective treatment could alleviate ongoing neurodegeneration, cell death and neurological impairment following TBI. Various neuroprotective drug candidates have been described, tested and proven effective in pre-clinical studies, including glutamate receptor antagonists, calcium-channel blockers, and caspase inhibitors. However, most of the scientific efforts have failed in translating the experimental results into clinical trials. Despite intensive research, effective neuroprotective therapies are lacking in the clinic, and TBI continues to be a major cause of morbidity and mortality. This paper provides an overview of the TBI pathophysiology leading to cell death and neurological impairment. We also discuss endogenously expressed neuroprotectants and drug candidates, which at this stage may still hold the potential for treating brain injured patients.


Subject(s)
Brain Injuries/drug therapy , Metallothionein/therapeutic use , Neuroprotective Agents/therapeutic use , Brain Injuries/physiopathology , Cell Death , Humans
16.
Prog Histochem Cytochem ; 44(1): 29-64, 2009.
Article in English | MEDLINE | ID: mdl-19348910

ABSTRACT

The antiapoptotic, antioxidant, proliferative, and angiogenic effects of metallothionein (MT)-I+II has resulted in increased focus on their role in oncogenesis, tumor progression, therapy response, and patient prognosis. Studies have reported increased expression of MT-I+II mRNA and protein in various human cancers; such as breast, kidney, lung, nasopharynx, ovary, prostate, salivary gland, testes, urinary bladder, cervical, endometrial, skin carcinoma, melanoma, acute lymphoblastic leukemia (ALL), and pancreatic cancers, where MT-I+II expression is sometimes correlated to higher tumor grade/stage, chemotherapy/radiation resistance, and poor prognosis. However, MT-I+II are downregulated in other types of tumors (e.g. hepatocellular, gastric, colorectal, central nervous system (CNS), and thyroid cancers) where MT-I+II is either inversely correlated or unrelated to mortality. Large discrepancies exist between different tumor types, and no distinct and reliable association exists between MT-I+II expression in tumor tissues and prognosis and therapy resistance. Furthermore, a parallel has been drawn between MT-I+II expression as a potential marker for prognosis, and MT-I+II's role as oncogenic factors, without any direct evidence supporting such a parallel. This review aims at discussing the role of MT-I+II both as a prognostic marker for survival and therapy response, as well as for the hypothesized role of MT-I+II as causal oncogenes.


Subject(s)
Biomarkers, Tumor , Metallothionein/physiology , Neoplasms/diagnosis , Neoplasms/physiopathology , Biomarkers, Tumor/chemistry , Humans , Metallothionein/chemistry , Metallothionein/classification , Neoplasms/mortality , Prognosis
17.
Histol Histopathol ; 24(5): 573-86, 2009 05.
Article in English | MEDLINE | ID: mdl-19283666

ABSTRACT

UNLABELLED: Traumatic brain injury represents a leading cause of morbidity in young individuals and there is an imperative need for neuroprotective treatments limiting the neurologic impairment following such injury. It has recently been demonstrated that bio-liberated gold ions liberated from small metallic gold implants reduce inflammation and neuronal apoptosis, while generating an increased neuronal stem cell response following focal brain damage. In this study mice were subjected to a unilateral traumatic cryo-lesion with concomitant injection of 25-45 microm gold particles near the lesion. Placebo-treated mice subjected to cryo-lesion served as controls. The effects of gold-treatment were investigated by examining gold-induced growth factor expression (VEGF and FGF) in the first two weeks after the insult, and the extent of the neurostimulatory effect of gold was explored by comparing cell proliferation in the subventricular zone as judged by immunohistochemical staining for CDC47. Vimentin staining revealed a decrease in activated microglia and a transient astrogliosis in response to the gold liberation. Moreover, gold ions significantly increase the expression of VEGF and FGF following trauma and a significant increase in cell proliferation in both the ipsilateral and the contralateral subventricular zone was found in response to gold-treatment. IN CONCLUSION: we confirmed the previously demonstrated anti-inflammatory effect of bio-liberated gold ions, and further show that metallic gold increases growth factor expression and adult neurogenesis.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/pathology , Fibroblast Growth Factors/metabolism , Gold/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Brain Injuries/metabolism , Cell Cycle Proteins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Female , Gold/administration & dosage , Immunohistochemistry , Inflammation/drug therapy , Inflammation/pathology , Mice , Mice, Inbred C57BL , Minichromosome Maintenance Complex Component 7 , Nerve Regeneration/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism
18.
Brain Res ; 1271: 103-13, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19328189

ABSTRACT

Brain injury represents a major health problem and may result in chronic inflammation and neurodegeneration. Due to antiinflammatory effects of gold, we have investigated the cerebral effects of metallic gold particles following a focal brain injury (freeze-lesion) in mice. Gold particles 20-45 microm in size or the vehicle (placebo) were implanted in the cortical tissue followed by a cortical freeze-lesioning. At 1-2 weeks post-injury, brains were analyzed by using immunohistochemistry and markers of inflammation, oxidative stress and apoptosis. This study shows that gold treatment significantly reduces the cerebral levels of tumor necrosis factor alpha (TNFalpha), oxidative DNA damage (as judged by 8-oxoguanine levels), and pro-apoptotic markers (cleaved caspase-3, cytochrome c leakage), when compared to those of controls. The data presented here points toward gold particles as a tool to modulate the cerebral response to injury.


Subject(s)
Apoptosis/drug effects , Brain Injuries/drug therapy , Gold/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/drug effects , Animals , Apoptosis/physiology , Biomarkers/analysis , Biomarkers/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Caspase 3/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytochromes c/metabolism , DNA Damage/drug effects , DNA Damage/physiology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Female , Gold/therapeutic use , Guanine/analogs & derivatives , Guanine/analysis , Guanine/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
19.
Leuk Lymphoma ; 50(2): 200-10, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19199157

ABSTRACT

Metallothionein (MT) expression is considered to be a prognostic factor that promotes tumor resistance to apoptosis. In non-Hodgkin lymphomas, MT is differentially expressed and constitutes a risk factor. We have characterised MT in lymph nodes of Hodgkin lymphoma (HL) [patients with nodular sclerosis (NSHL), mixed cellularity (MCHL), lymphocyte-rich classical HL (LRCHL) and nodular lymphocyte predominant HL (NLPHL)] and in controls. MT expression is significantly and differentially altered in the HL subtypes. NSHL and MCHL show highly increased MT throughout the lymph node. In contrast, MT is barely increased in LRCHL relative to controls. NLPHL shows a distinct pattern of heterogeneous MT with increased MT in nodular areas surrounded by MT-negative tissue. The cellular MT sources are reactive, infiltrating (non-neoplastic) cells, whereas neoplastic cells are devoid of MT. We show for the first time that MT is differentially expressed in subclassified HL.


Subject(s)
Biomarkers, Tumor/metabolism , Hodgkin Disease/classification , Hodgkin Disease/metabolism , Metallothionein/metabolism , Biopsy , Hodgkin Disease/pathology , Hodgkin Disease/therapy , Humans , Treatment Outcome
20.
Arthritis Res Ther ; 10(4): R76, 2008.
Article in English | MEDLINE | ID: mdl-18601746

ABSTRACT

INTRODUCTION: Metallothionein (MT) isoforms I + II are polypeptides with potent antioxidative and anti-inflammatory properties. In healthy kidneys, MT-I+II have been described as intracellular proteins of proximal tubular cells. The aim of the present study was to investigate whether the renal MT-I+II expression profile is altered during lupus nephritis. METHODS: Immunohistochemistry was performed on renal biopsies from 37 patients with lupus nephritis. Four specimens of healthy renal tissue served as controls. Clinicopathological correlation studies and renal survival analyses were performed by means of standard statistical methods. RESULTS: Proximal tubules displaying epithelial cell MT-I+II depletion in combination with luminal MT-I+II expression were observed in 31 out of 37 of the lupus nephritis specimens, but not in any of the control sections (P = 0.006). The tubular MT score, defined as the median number of proximal tubules displaying this MT expression pattern per high-power microscope field (40x magnification), was positively correlated to the creatinine clearance in the lupus nephritis cohort (P = 0.01). Furthermore, a tubular MT score below the median value of the cohort emerged as a significant predictor of a poor renal outcome in renal survival analyses. Thus, patients with a tubular MT score < 1.0 had a 6.2-times higher risk of developing end-stage renal disease than patients with a tubular MT score >or= 1.0 (P = 0.03). CONCLUSION: Lupus nephritis is associated with significant alterations in renal MT-I+II expression. Our data indicate that important prognostic information can be deduced from the renal MT-I+II expression profile in systemic lupus erythematosus patients with nephritis.


Subject(s)
Kidney/metabolism , Lupus Nephritis/metabolism , Metallothionein/metabolism , Adult , Biopsy , Case-Control Studies , Female , Gene Expression Profiling , Glomerulonephritis/diagnosis , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Humans , Kidney/pathology , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Longitudinal Studies , Lupus Nephritis/diagnosis , Lupus Nephritis/pathology , Male , Metallothionein/genetics , Middle Aged , Prognosis , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...