Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Eur J Neurosci ; 52(4): 3223-3241, 2020 08.
Article in English | MEDLINE | ID: mdl-31954073

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, characterised by the degeneration of motor neurons innervating skeletal muscle. The mechanisms underlying neurodegeneration in ALS are not yet fully elucidated, and with current therapeutics only able to extend lifespan by a matter of months there is a clear need for novel therapies to increase lifespan and patient quality of life. Here, we evaluated whether moderate-intensity treadmill exercise and/or treatment with metallothionein-2 (MT2), a neuroprotective protein, could improve survival, behavioural or neuropathological outcomes in SOD1G93A familial ALS mice. Six-week-old female SOD1G93A mice were allocated to one of four treatment groups: MT2 injection, i.m.; moderate treadmill exercise; neither MT2 nor exercise; or both MT2 and exercise. MT2-treated mice survived around 3% longer than vehicle-treated mice, with this mild effect reaching statistical significance in Cox proportional hazards analysis once adjusted for potential confounders. Mixed model body weight trajectories over time indicated that MT2-treated mice, with or without exercise, reached maximum body weight at a later age, suggesting a delay in disease onset of around 4% compared to saline-treated mice. Exercise alone did not significantly increase survival or delay disease onset, and neither exercise nor MT2 substantially ameliorated gait abnormalities or muscle strength loss. We conclude that neither exercise nor MT2 treatment was detrimental in female SOD1G93A mice, and further study could determine whether the mild effect of peripheral MT2 administration on disease onset and survival could be improved via direct administration of MT2 to the central nervous system.


Subject(s)
Amyotrophic Lateral Sclerosis , Metallothionein/therapeutic use , Physical Conditioning, Animal , Superoxide Dismutase-1 , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Disease Models, Animal , Female , Mice , Mice, Transgenic , Quality of Life , Superoxide Dismutase-1/genetics
2.
J Hematol Oncol ; 11(1): 107, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30139373

ABSTRACT

Metallothioneins (MTs) are small cysteine-rich proteins that play important roles in metal homeostasis and protection against heavy metal toxicity, DNA damage, and oxidative stress. In humans, MTs have four main isoforms (MT1, MT2, MT3, and MT4) that are encoded by genes located on chromosome 16q13. MT1 comprises eight known functional (sub)isoforms (MT1A, MT1B, MT1E, MT1F, MT1G, MT1H, MT1M, and MT1X). Emerging evidence shows that MTs play a pivotal role in tumor formation, progression, and drug resistance. However, the expression of MTs is not universal in all human tumors and may depend on the type and differentiation status of tumors, as well as other environmental stimuli or gene mutations. More importantly, the differential expression of particular MT isoforms can be utilized for tumor diagnosis and therapy. This review summarizes the recent knowledge on the functions and mechanisms of MTs in carcinogenesis and describes the differential expression and regulation of MT isoforms in various malignant tumors. The roles of MTs in tumor growth, differentiation, angiogenesis, metastasis, microenvironment remodeling, immune escape, and drug resistance are also discussed. Finally, this review highlights the potential of MTs as biomarkers for cancer diagnosis and prognosis and introduces some current applications of targeting MT isoforms in cancer therapy. The knowledge on the MTs may provide new insights for treating cancer and bring hope for the elimination of cancer.


Subject(s)
Carcinogenesis/metabolism , Metallothionein/therapeutic use , Neoplasms/metabolism , Humans , Metallothionein/pharmacology
3.
Exp Mol Med ; 50(8): 1-13, 2018 08 17.
Article in English | MEDLINE | ID: mdl-30120245

ABSTRACT

An excess of reactive oxygen species (ROS) relative to the antioxidant capacity causes oxidative stress, which plays a role in the development of Parkinson's disease (PD). Because mitochondria are both sites of ROS generation and targets of ROS damage, the delivery of antioxidants to mitochondria might prevent or alleviate PD. To transduce the antioxidant protein human metallothionein 1A (hMT1A) into mitochondria, we computationally designed a cell-penetrating artificial mitochondria-targeting peptide (CAMP). The recombinant CAMP-conjugated hMT1A fusion protein (CAMP-hMT1A) successfully localized to the mitochondria. Treating a cell culture model of PD with CAMP-hMT1A restored tyrosine hydroxylase expression and mitochondrial activity and reduced ROS production. Furthermore, injection of CAMP-hMT1A into the brain of a mouse model of PD rescued movement impairment and dopaminergic neuronal degeneration. CAMP-hMT1A delivery into mitochondria might be therapeutic against PD by alleviating mitochondrial damage, and we predict that CAMP could be used to deliver other cargo proteins to the mitochondria.


Subject(s)
Cell-Penetrating Peptides/therapeutic use , Metallothionein/therapeutic use , Mitochondria/metabolism , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Amino Acid Sequence , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Computer Simulation , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Humans , Metallothionein/pharmacology , Mice , Mitochondria/drug effects , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/pathology , Protein Transport , Recombinant Fusion Proteins/therapeutic use , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
4.
Curr Pharm Des ; 24(27): 3155-3161, 2018.
Article in English | MEDLINE | ID: mdl-30014800

ABSTRACT

Inflammatory bowel disease (IBD) is a group of disorders characterized by chronic inflammation within the gastrointestinal tract. It is a multifactorial disease associated with immune-cell mediated oxidative damage to the intestinal mucosa. There is no cure for IBD, but anti-cytokine therapy can limit target inflammation and disease progression. Unfortunately, many patients are nonresponsive or develop resistance to anti-cytokine therapy over time creating a need for new therapeutic agents. Metallothionein (MT) is a small, highly conserved stress response protein that has been shown to modulate the immune response as a pro-inflammatory agent, regulates divalent heavy metal homeostasis, and acts as a reactive metabolite scavenger. Our research, as well as other groups studying MT, has described MT induction and release during IBD inflammatory stress response. The release of MT results in activation of inflammatory responses leading to progressive inflammation and subsequent expansion of MT synthesis. A monoclonal antibody specific for MT has been used in murine models of IBD and should only target the extracellular pool of MT, thus representing a novel therapeutic approach to this disease.


Subject(s)
Inflammatory Bowel Diseases/therapy , Metallothionein/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Humans , Immunotherapy , Inflammatory Bowel Diseases/immunology
5.
Curr Pharm Des ; 23(33): 5001-5009, 2017.
Article in English | MEDLINE | ID: mdl-28641540

ABSTRACT

Lou Gehrig's disease, a synonym of amyotrophic lateral sclerosis, is an adult-onset lethal neurodegenerative disorder. Irrespective of extensive efforts to elucidate the pathogenesis of the disease and searches for therapies, no favorable pharmacotherapeutic strategies have yet to be proposed. In a popular rodent model of ALS, G93A SOD1 strain of mouse, intracellular copper conditions were geared toward copper accumulation inside cells, resulting in an acceleration of oxidative stress and apoptotic process. Disruption of intracellular copper homeostasis was common to transgenic mice expressing human mutant SOD1s. In this review, the novel hypothesis that disruption of intracellular copper homeostasis could be involved in the development of the disease was introduced. Based upon the hypothesis, therapeutic outcomes of agents that are capable of correcting and/or modifying intracellular copper homeostasis are described. Administration of ammonium tetrathiomolybdate, a selective intracellular copper chelator, delayed onset, slowed progression, and prolonged survival of a rodent model of the disease (G93A SOD1 mice). Metallothionein is a low molecular weight, cysteine-rich, metal-binding cytoplasmic protein that has beneficial properties in detoxification of toxic heavy metals, homeostatic regulation of intracellular essential trace elements, including copper, antioxidant, and antiapoptotic roles. In animal experiments of the G93A SOD1 mice, an increase of metallothionein proteins by means of induction by exercise or dexamethasone, genetic overexpression, or intraperitoneal administration, all resulted in a preferable outcome. The therapeutic effects were not inferior to those of approved drugs for ALS in humans. These observations suggest that metallothionein could be worth investigating the therapeutic potential in clinical use.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Metallothionein/metabolism , Metallothionein/therapeutic use , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Chelating Agents/metabolism , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Humans , Metallothionein/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology
6.
J Biol Chem ; 291(3): 1092-102, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26598525

ABSTRACT

The low-density lipoprotein receptor-related protein receptors 1 and 2 (LRP1 and LRP2) are emerging as important cell signaling mediators in modulating neuronal growth and repair. We examined whether LRP1 and LRP2 are able to mediate a specific aspect of neuronal growth: axon guidance. We sought to identify LRP1 and LRP2 ligands that could induce axonal chemoattraction, which might have therapeutic potential. Using embryonic sensory neurons (rat dorsal root ganglia) in a growth cone turning assay, we tested a range of LRP1 and LRP2 ligands for the ability to guide growth cone navigation. Three ligands were chemorepulsive: α-2-macroglobulin, tissue plasminogen activator, and metallothionein III. Conversely, only one LRP ligand, metallothionein II, was found to be chemoattractive. Chemoattraction toward a gradient of metallothionein II was calcium-dependent, required the expression of both LRP1 and LRP2, and likely involves further co-receptors such as the tropomyosin-related kinase A (TrkA) receptor. The potential for LRP-mediated chemoattraction to mediate axonal regeneration was examined in vivo in a model of chemical denervation in adult rats. In these in vivo studies, metallothionein II was shown to enhance epidermal nerve fiber regeneration so that it was complete within 7 days compared with 14 days in saline-treated animals. Our data demonstrate that both LRP1 and LRP2 are necessary for metallothionein II-mediated chemotactic signal transduction and that they may form part of a signaling complex. Furthermore, the data suggest that LRP-mediated chemoattraction represents a novel, non-classical signaling system that has therapeutic potential as a disease-modifying agent for the injured peripheral nervous system.


Subject(s)
Axons/physiology , Low Density Lipoprotein Receptor-Related Protein-1/agonists , Low Density Lipoprotein Receptor-Related Protein-2/agonists , Nerve Regeneration , Nerve Tissue Proteins/agonists , Neurogenesis , Peripheral Nerves/physiology , Animals , Axons/drug effects , Calcium Signaling/drug effects , Cells, Cultured , Chemotaxis/drug effects , Epidermis/drug effects , Epidermis/innervation , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Growth Cones/drug effects , Growth Cones/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Metallothionein/pharmacology , Metallothionein/therapeutic use , Nerve Regeneration/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Peripheral Nerves/cytology , Peripheral Nerves/drug effects , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/physiopathology , RNA Interference , Rabbits , Rats, Sprague-Dawley
7.
PLoS One ; 10(12): e0144035, 2015.
Article in English | MEDLINE | ID: mdl-26658636

ABSTRACT

Metallothionein-II (MT-II) is an ubiquitously expressed small-molecular-weight protein and highly induced in various species and tissues upon stress, inflammation, and ischemia. MT-deficiency exacerbates ischemic injury in rodent stroke models in vitro and in vivo. However, there is conflicting data on the potential neuroprotective effect of exogenously applied metallothionein. Thus, we applied MT-II in an in vitro stroke model and intraperitoneally (i.p.) in two in vivo standard models of transient middle cerebral artery occlusion (MCAO) (a 'stringent' one [60 min MCAO/48 h reperfusion] and a 'mild' one [30 min MCAO/72 h reperfusion]), as well as i.v. together with recombinant tissue plasminogen activator (rtPA) to evaluate if exogenous MT-II-application protects against ischemic stroke. Whereas MT-II did not protect against 60 min MCAO, there was a significant reduction of direct and indirect infarct volumes and neurological deficit in the MT-II (i.p.) treated animals in the 'mild' model at 3d after MCAO. Furthermore, MT-II also improved survival of the mice after MCAO, suppressed TNF-α mRNA induction in ischemic brain tissue, and protected primary neuronal cells against oxygen-glucose-deprivation in vitro. Thus, exogenous application of MT-II protects against ischemic injury in vitro and in vivo. However, long-term studies with different species and larger sampling sizes are required before a clinical use can be envisaged.


Subject(s)
Brain Ischemia/drug therapy , Metallothionein/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Cells, Cultured , Cytokines/genetics , In Vitro Techniques , Inflammation Mediators/metabolism , Male , Metallothionein/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neuroprotective Agents/pharmacology
8.
Free Radic Res ; 49(10): 1187-98, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25968954

ABSTRACT

AIMS: Endoplasmic reticulum (ER) stress exerts myocardial oxidative stress, apoptosis, and contractile anomalies, although the precise interplay between ER stress and apoptosis remains elusive. This study was designed to examine the impact of the cysteine-rich free radical scavenger metallothionein on ER stress-induced myocardial contractile defect and underlying mechanisms. METHODS AND RESULTS: Wild-type friendly virus B and transgenic mice with cardiac-specific overexpression of metallothionein were challenged with the ER stress inducer tunicamycin (1 mg/kg, intraperitoneal, 48 h) prior to the assessment of myocardial function, oxidative stress, and apoptosis. Our results revealed that tunicamycin promoted cardiac remodeling (enlarged left ventricular end systolic/diastolic diameters with little changes in left ventricular wall thickness), suppressed fractional shortening and cardiomyocyte contractile function, elevated resting Ca(2+), decreased stimulated Ca(2+) release, prolonged intracellular Ca(2+) clearance, and downregulated sarco(endo)plasmic reticulum Ca(2+)-ATPase levels, the effects of which were negated by metallothionein. Treatment with tunicamycin caused cardiomyocyte mitochondrial injury, as evidenced by decreased mitochondrial membrane potential (∆Ñ°m, assessed by JC-1 staining), the effect of which was negated by the antioxidant. Moreover, tunicamycin challenge dramatically facilitated myocardial apoptosis as manifested by increased Bax, caspase 9, and caspase 12 protein levels, as well as elevated caspase 3 activity. Interestingly, metallothionein transgene significantly alleviated tunicamycin-induced myocardial apoptosis. CONCLUSION: Taken together, our data favor a beneficial effect of metallothionein against ER stress-induced cardiac dysfunction possibly associated with attenuation of myocardial apoptosis.


Subject(s)
Antioxidants/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Free Radical Scavengers/therapeutic use , Heart/drug effects , Metallothionein/therapeutic use , Myocardial Contraction/drug effects , Myocardium/pathology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Biomarkers , Calcium Signaling/drug effects , Calcium-Transporting ATPases/metabolism , Free Radical Scavengers/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Metallothionein/pharmacology , Mice , Mice, Transgenic , Tunicamycin/pharmacology , Tunicamycin/therapeutic use , Ventricular Remodeling/drug effects
9.
Oxid Med Cell Longev ; 2014: 436429, 2014.
Article in English | MEDLINE | ID: mdl-24719677

ABSTRACT

After transient cerebral ischemia and reperfusion (I/R), damaging mechanisms, such as excitotoxicity and oxidative stress, lead to irreversible neurological deficits. The induction of metallothionein-II (MT-II) protein is an endogenous mechanism after I/R. Our aim was to evaluate the neuroprotective effect of MT-II after I/R in rats. Male Wistar rats were transiently occluded at the middle cerebral artery for 2 h, followed by reperfusion. Rats received either MT (10 µg per rat i.p.) or vehicle after ischemia. Lipid peroxidation (LP) was measured 22 h after reperfusion in frontal cortex and hippocampus; also, neurological deficit was evaluated after ischemia, using the Longa scoring scale. Infarction area was analyzed 72 hours after ischemia. Results showed increased LP in frontal cortex (30.7%) and hippocampus (26.4%), as compared to control group; this effect was fully reversed by MT treatment. Likewise, we also observed a diminished neurological deficit assessed by the Longa scale in those animals treated with MT compared to control group values. The MT-treated group showed a significant (P < 0.05) reduction of 39.9% in the infarction area, only at the level of hippocampus, as compared to control group. Results suggest that MT-II may be a novel neuroprotective treatment to prevent ischemia injury.


Subject(s)
Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/physiopathology , Lipid Peroxidation/drug effects , Metallothionein/therapeutic use , Recovery of Function/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Animals , Frontal Lobe/drug effects , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/pathology , Male , Metallothionein/administration & dosage , Metallothionein/pharmacology , Rabbits , Rats , Rats, Wistar , Reperfusion Injury/complications , Reperfusion Injury/pathology
10.
Mini Rev Med Chem ; 14(3): 271-81, 2014.
Article in English | MEDLINE | ID: mdl-24456271

ABSTRACT

Alzheimer's disease (AD) is characterized by the deposition of amyloid-ß (Aß) peptide in the brains of AD patients. Such a process is linked to the binding of metal ions (e.g., Cu, Fe and Zn) with Aß. As a result, metal chelation could be used as a rational therapeutic pathway for the treatment of AD. In this review, we address some noteworthy advances on the utilization of metal chelators, such as native metallothioneins and synthetic compounds, as potential therapeutic agents for AD. In addition, the future design and utility of metal chelating drugs as well as the strategy pursued to transport metal chelators into the brain are highlighted. We believe that this contribution will be valuable for the design of metal-chelating drugs for AD treatment.


Subject(s)
Amyloid beta-Peptides/metabolism , Chelating Agents/chemical synthesis , Chelating Agents/pharmacology , Metallothionein/pharmacology , Metals/isolation & purification , Metals/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Animals , Biological Transport , Chelating Agents/metabolism , Chelating Agents/therapeutic use , Humans , Metallothionein/chemistry , Metallothionein/metabolism , Metallothionein/therapeutic use , Molecular Sequence Data
11.
J Control Release ; 172(3): 1092-101, 2013 Dec 28.
Article in English | MEDLINE | ID: mdl-24103814

ABSTRACT

Subcutaneous site is ideal for clinical islet transplantation because it has the advantage of simple operation procedure under local anesthesia and can be biopsied when needed. However, the transplantation outcomes at subcutaneous site have been disappointing due to hypoxia-induced oxidative stress by poor vascularization. We hypothesized that subcutaneously transplanted islets would have hypoxia resistance by using internalization of metallothionein (MT), an antioxidant scavenging enzyme, which was mediated by fusion between MT and cell penetrating Tat peptide. The Tat-MT was dose-dependently transduced into islets without any damage. Tat-MT-treated islets could be protected from oxidative stress induced by intracellular nitric oxide donor, sodium nitroprusside (SNP). When Tat-MT-treated islets were subcutaneously transplanted into diabetic nude mice, they normally controlled the blood glucose levels without severe fluctuation (median survival time (MST): >30 days), whereas most untreated islets were rejected (MST 17 days). From the intraperitoneal glucose tolerance test 5 days after posttransplantation, glucose responsiveness of Tat-MT-treated islets was similar to that of normal healthy mice, while untreated islets had delayed glucose responsiveness. From the results of immunohistochemical stain, Tat-MT-treated islets had strong anti-insulin positive cells and lower anti-HIF-1α positive cells. However, untreated islets had rare anti-insulin positive cells and strong anti-HIF-1α-positive cells. Collectively, these findings demonstrated that Tat-MT delivery into islet could offer a new strategy for successful islet transplantation under subcutaneous space.


Subject(s)
Antioxidants/therapeutic use , Gene Products, tat/therapeutic use , Hypoxia/prevention & control , Islets of Langerhans Transplantation/methods , Metallothionein/therapeutic use , Amino Acid Sequence , Animals , Antioxidants/administration & dosage , Antioxidants/chemistry , Gene Products, tat/administration & dosage , Gene Products, tat/chemistry , Glucose/metabolism , Glucose Tolerance Test , Humans , Hypoxia/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Metallothionein/administration & dosage , Metallothionein/chemistry , Mice , Molecular Sequence Data , Oxidative Stress , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/therapeutic use
12.
Biol Trace Elem Res ; 153(1-3): 291-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23625696

ABSTRACT

Zinc exerts a wide range of important biological roles. The present study was carried out to investigate the effects of zinc threoninate chelate in blood glucose levels, lipid peroxidation, activities of antioxidant defense systems and nitrite concentration, and histology of the pancreas in diabetic rats. Wistar rats were intravenously injected with a single dose of streptozotocin to induce diabetes. Then, diabetic rats were administrated orally with zinc threoninate chelate (3, 6, and 9 mg/kg body weight) once daily for 7 weeks. Fasting blood glucose was monitored weekly. At the end of the experimental period, the diabetic rats were killed, and levels of serum insulin, malondialdehyde, and nitric oxide, activities of glutathione peroxidase, total superoxide dismutase, copper/zinc-superoxide dismutase, and nitric oxide synthase were determined; pancreas was examined histopathologically as well. Zinc threoninate chelate significantly reduced the blood glucose levels and significantly increased the serum insulin levels in diabetic rats. In addition, zinc threoninate chelate caused a significant increase in activities of antioxidant enzymes and significant decrease in nitrite concentration and malondialdehyde formation in the pancreas and serum of diabetic rats. These biochemical observations were supplemented by histopathological examination of the pancreas. These results suggested that the antidiabetic effect of zinc threoninate chelate may be related to its antioxidative stress ability in diabetic rats.


Subject(s)
Antioxidants/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Metallothionein/pharmacology , Oxidative Stress/drug effects , Pancreas/drug effects , Animals , Antioxidants/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Male , Metallothionein/therapeutic use , Pancreas/metabolism , Rats , Rats, Wistar , Streptozocin
13.
Curr Pharm Biotechnol ; 14(4): 408-13, 2013.
Article in English | MEDLINE | ID: mdl-23590138

ABSTRACT

Metallothionein (MT) is a small molecular and multi-functional protein containing four atoms of copper (Cu) and three atoms of zinc (Zn) per molecule. It was isolated from the horse kidney in 1957 and half a century has passed since then. Although MT was found to work as a modulator of Zn and induce anti-oxidant reaction, the precise functions and its functional mechanisms remain to be elucidated. Over the years, a new isoform of MT, MT-III (also called growth inhibitory factor (GIF)), has been found in the brain, which was markedly diminished in the brain of Alzheimer's disease (AD). Many new findings on MT have been discovered in neurodegenerative diseases other than AD such as amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), prion disease, brain trauma, brain ischemia, and psychiatric diseases. In ALS in particular, MTs were markedly diminished in the spinal cord of patients with ALS. Initially, MT, which easily binds to cadmium (Cd) and copper (Cu), was considered to be toxic to our bodies. Molecular biological technologies enabled the production of recombinant MT saturated with zinc (Zn). MT has a high potential for the treatment of neurodegenerative diseases such as ALS, AD, and PD owing to its various functions including anti-oxidant properties and modulators not only for Zn but for Cu in the extra- and intracellular spaces. On the other hand, there are still various problems on MT to be elucidated in detail, including their binding proteins and functional mechanisms.


Subject(s)
Metallothionein/pharmacology , Metallothionein/therapeutic use , Neurodegenerative Diseases/drug therapy , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/drug effects , Brain/metabolism , Humans , Neurodegenerative Diseases/metabolism
14.
J Control Release ; 169(3): 306-12, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-23391442

ABSTRACT

Ischemic heart diseases caused by insufficient oxygen supply to the cardiac muscle require pharmaceutical agents for the prevention of the progress and recurrence. Metallothionein (MT) has a potential as a protein therapeutic for the treatment of this disease due to its anti-oxidative effects under stressful conditions. In spite of its therapeutic potential, efficient delivery systems need to be developed to overcome limitations such as low transduction efficiency, instability and short half-life in the body. To enhance intra-cellular transduction efficiency, Tat sequence as a protein transduction domain (PTD) was fused with MT in a recombinant method. Anti-apoptotic and anti-oxidative effects of Tat-MT fusion protein were evaluated under hyperglycemia and hypoxia stress conditions in cultured H9c2 cells. Recovery of cardiac functions by anti-apoptotic and anti-fibrotic effects of Tat-MT was confirmed in an ischemia/reperfusion (I/R) rat myocardial infarction model. Tat-MT fusion protein effectively protected H9c2 cells under stressful conditions by reducing intracellular ROS production and inhibiting caspase-3 activation. Tat-MT fusion protein inhibited apoptosis, reduced fibrosis area and enhanced cardiac functions in I/R. Tat-MT fusion protein could be a promising therapeutic for the treatment of ischemic heart diseases.


Subject(s)
Metallothionein/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Animals , Apoptosis/drug effects , Cell Line , Gene Products, tat/genetics , Gene Products, tat/pharmacology , Gene Products, tat/therapeutic use , Hyperglycemia/prevention & control , Male , Metallothionein/genetics , Metallothionein/pharmacology , Mice , Oxidative Stress/drug effects , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
15.
Neurosci Lett ; 514(1): 102-5, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22405890

ABSTRACT

After spinal cord injury (SCI), a complex cascade of pathophysiological processes rapidly damages the nervous tissue. The initial damage spreads to the surrounding tissue by different mechanisms, including oxidative stress. We have recently reported that the induction of metallothionein (MT) protein is an endogenous rapid-response mechanism after SCI. Since the participation of MT in neuroprotective processes after SCI is still unknown, the aim of the present study was to evaluate the possible neuroprotective effect of exogenously administered MT-II during the acute phase after SCI in rats. Female Wistar rats weighing 200-250g were submitted to spinal cord contusion by means of a computer-controlled device (NYU impactor). Rats received several doses of MT-II (3.2, 10 and 100µg) at 2 and 8h after SCI. Results of the BBB scale were statistically analysed using an ANOVA of repeated-measures, followed by Tukey's test. Among the three doses tested, only 10 and 100µg were able to significantly increase (p<0.05) BBB scale scores eight weeks after SCI from a mean of 7.88 in the control group, to means of 12.63 and 10.88 for the 10 and 100µg doses of MT-II, respectively. The amount of spared tissue was also higher in the groups treated with 10 and 100µg, as compared to the control group values. Results from the present study demonstrate a significant neuroprotective effect of exogenously administered MT-II. Further studies are needed in order to characterize the mechanisms involved in this neuroprotective action.


Subject(s)
Metallothionein/pharmacology , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Animals , Female , Metallothionein/therapeutic use , Motor Activity/physiology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar , Recovery of Function/physiology , Spinal Cord/physiopathology , Spinal Cord Injuries/physiopathology
16.
Hemoglobin ; 33(5): 370-7, 2009.
Article in English | MEDLINE | ID: mdl-19821780

ABSTRACT

Oxidative stress is considered to be the main cause for several chronic diseases including diabetes. Through hyperglycemia, hyperlipidemia, hypertension and possible iron dyshomeostasis, diabetes induces oxidative stress that causes damage to multiple organs, leading to various complications. Therefore, antioxidant therapy may be an interesting approach to prevent diabetes and diabetic complications. Metallothionein as a potent antioxidant was found to significantly protect heart and kidney against diabetes-induced pathophysiological changes. Zinc as an important trace element and a metallothionein inducer was found to have same protective function. Since diabetes would impair defensive system, including growth factor reduction, exogenous supplementation of fibroblast growth factor (FGF) significantly prevented diabetes-induced cardiac oxidative damage and wound healing impairment. These studies suggest that protective agents such as metallothionein, zinc and FGFs play an important role in preventing the development of diabetes and diabetic complications.


Subject(s)
Diabetes Complications/prevention & control , Diabetes Mellitus/drug therapy , Diabetes Mellitus/etiology , Oxidative Stress/drug effects , Animals , Antioxidants/therapeutic use , Disease Models, Animal , Fibroblast Growth Factors/therapeutic use , Humans , Iron Overload/drug therapy , Iron Overload/etiology , Metallothionein/therapeutic use , Trace Elements/therapeutic use , Zinc/therapeutic use
17.
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
18.
Diabetes ; 58(6): 1391-402, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19324938

ABSTRACT

OBJECTIVE: Glycogen synthase kinase (GSK)-3beta plays an important role in cardiomyopathies. Cardiac-specific metallothionein-overexpressing transgenic (MT-TG) mice were highly resistant to diabetes-induced cardiomyopathy. Therefore, we investigated whether metallothionein cardiac protection against diabetes is mediated by inactivation of GSK-3beta. RESEARCH DESIGN AND METHODS: Diabetes was induced with streptozotocin in both MT-TG and wild-type mice. Changes of energy metabolism-related molecules, lipid accumulation, inflammation, nitrosative damage, and fibrotic remodeling were examined in the hearts of diabetic mice 2 weeks, 2 months, and 5 months after the onset of diabetes with Western blotting, RT-PCR, and immunohistochemical assays. RESULTS: Activation (dephosphorylation) of GSK-3beta was evidenced in the hearts of wild-type diabetic mice but not MT-TG diabetic mice. Correspondingly, cardiac glycogen synthase phosphorylation, hexokinase II, PPARalpha, and PGC-1alpha expression, which mediate glucose and lipid metabolisms, were significantly changed along with cardiac lipid accumulation, inflammation (TNF-alpha, plasminogen activator inhibitor 1 [PAI-1], and intracellular adhesion molecule 1 [ICAM-1]), nitrosative damage (3-nitrotyrosin accumulation), and fibrosis in the wild-type diabetic mice. The above pathological changes were completely prevented either by cardiac metallothionein in the MT-TG diabetic mice or by inhibition of GSK-3beta activity in the wild-type diabetic mice with a GSK-3beta-specific inhibitor. CONCLUSIONS: These results suggest that activation of GSK-3beta plays a critical role in diabetes-related changes in cardiac energy metabolism, inflammation, nitrosative damage, and remodeling. Metallothionein inactivation of GSK-3beta plays a critical role in preventing diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/prevention & control , Glycogen Synthase Kinase 3/antagonists & inhibitors , Inflammation/prevention & control , Metallothionein/genetics , Metallothionein/pharmacology , Myocardium/metabolism , Ventricular Remodeling/genetics , Animals , Diabetic Angiopathies/pathology , Energy Metabolism , Glycogen Synthase Kinase 3 beta , Metallothionein/therapeutic use , Mice , Mice, Inbred Strains , Mice, Transgenic
19.
Belo Horizonte; s.n; 2009. 157 p. ilus, tab.
Thesis in Portuguese | LILACS, BBO - Dentistry | ID: lil-557493

ABSTRACT

Cistos e tumores odontogênicos são lesões originadas dos tecidos que formam os dentes e apresentam diferentes comportamentos biológicos. A metalotioneíra (MT) é relacionada à homeostase de metais, regulação da diferenciação e proliferação celular e inibição da apoptose. Com relação aos cistos e tumores odontogênicos, a MT poderia ter um papel na regulação da diferenciação e proliferação celuar e na inibição da apoptose, refletindo no comportamento biológico. Os objetivos são avaliar e comparar a expressão da MT entre: 1) cistos odontogêncios e tumor odontogênico ceratocístico (TOC); 2) TOC associados à Síndrome do Carcinoma Basocelular Nevóide (SCBN) e não associados; 3) tumores odontogênicos benignos. Objetivou-se também correlacionar a imuno-expressão da MT com a proliferação celular e com a inflamação. A amostra incluiu cisto radicular (CR), cisto dentígero (CD), TOC (primário associado ou não à (SCBN), cisto odontogênico ortoceratinizado (COO), ameloblastoma sólido (ABS), tumor odontogênico escamoso (TOE), tumor odontogênico adenomatóide (TOA), tumor odontogênico cístico calcificante (TOCC) e tumor odontogênico epitelial calcificante (TOEC). Foi realizada imunoistoquímica para MT, Ki-67 e PCNA...


Subject(s)
Humans , Male , Female , Ameloblastoma/physiopathology , Odontogenic Cysts/physiopathology , Metallothionein/therapeutic use , Odontogenic Tumors/physiopathology , /therapeutic use , Immunohistochemistry , Basal Cell Nevus Syndrome/therapy
20.
Crit Care Med ; 36(7): 2106-16, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18552690

ABSTRACT

OBJECTIVE: Antioxidant therapy has shown some promise in critical care medicine in which glutathione depletion and heart failure are often seen in critically ill patients. This study was designed to examine the impact of glutathione depletion and the free radical scavenger, metallothionein (MT), on cardiac function. DESIGN: Friend virus B and MT transgenic mice were given the glutathione synthase inhibitor buthionine sulfoximine (buthionine sulfoximine [BSO], 30 mmol/L) in drinking water for 2 wks. MEASUREMENTS: Echocardiographic and cardiomyocyte functions were evaluated, including myocardial geometry, fraction shortening, peak shortening, time-to-90% relengthening (TR90), maximal velocity of shortening/relengthening (+/-dL/dt), intracellular Ca2+ rise, sarcoplasmic reticulum Ca2+ release, and intracellular Ca2+ decay rate. Sacro (endo)plasmic reticulum Ca2+-ATPase function was evaluated by 45Ca uptake. Highly reactive oxygen species, caspase-3, and aconitase activity were detected by fluorescent probe and colorimetric assays. MAIN RESULT: BSO elicited lipid peroxidation, protein carbonyl formation, mitochondrial damage, and apoptosis. BSO also reduced wall thickness, enhanced end systolic diameter, depressed fraction shortening, peak shortening, +/-dL/dt, sarcoplasmic reticulum Ca2+ release, 45Ca uptake, and intracellular Ca2+ decay, leading to prolonged TR90. BSO-induced mitochondrial loss and myofilament aberration. MT transgene itself had little effect on myocardial mechanics and ultrastructure. However, it alleviated BSO-induced myocardial functional, morphologic, and carbonyl changes. Western blot analysis showed reduced expression of sacro (endo)plasmic reticulum Ca2+-ATPase2a, Bcl-2 and phosphorylated GSK-3beta, enhanced calreticulin, Bax, p53, myosin heavy chain-beta isozyme switch, and IkappaB phosphorylation in FVB-BSO mice, all of which with the exception of p53 were nullified by MT. CONCLUSION: Our findings suggest a pathologic role of glutathione depletion in cardiac dysfunction and the therapeutic potential of antioxidants.


Subject(s)
Buthionine Sulfoximine/pharmacology , Cardiomyopathies/drug therapy , Enzyme Inhibitors/pharmacology , Glutathione/deficiency , Metallothionein/therapeutic use , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/etiology , Glutathione Synthase/antagonists & inhibitors , Lipid Peroxidation/drug effects , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL