Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Microb Cell Fact ; 22(1): 44, 2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36879280

ABSTRACT

BACKGROUND: Micafungin is an echinocandin-type antifungal agent used for the clinical treatment of invasive fungal infections. It is semisynthesized from the sulfonated lipohexapeptide FR901379, a nonribosomal peptide produced by the filamentous fungus Coleophoma empetri. However, the low fermentation efficiency of FR901379 increases the cost of micafungin production and hinders its widespread clinical application. RESULTS: Here, a highly efficient FR901379-producing strain was constructed via systems metabolic engineering in C. empetri MEFC09. First, the biosynthesis pathway of FR901379 was optimized by overexpressing the rate-limiting enzymes cytochrome P450 McfF and McfH, which successfully eliminated the accumulation of unwanted byproducts and increased the production of FR901379. Then, the functions of putative self-resistance genes encoding ß-1,3-glucan synthase were evaluated in vivo. The deletion of CEfks1 affected growth and resulted in more spherical cells. Additionally, the transcriptional activator McfJ for the regulation of FR901379 biosynthesis was identified and applied in metabolic engineering. Overexpressing mcfJ markedly increased the production of FR901379 from 0.3 g/L to 1.3 g/L. Finally, the engineered strain coexpressing mcfJ, mcfF, and mcfH was constructed for additive effects, and the FR901379 titer reached 4.0 g/L under fed-batch conditions in a 5 L bioreactor. CONCLUSIONS: This study represents a significant improvement for the production of FR901379 and provides guidance for the establishment of efficient fungal cell factories for other echinocandins.


Subject(s)
Alkanesulfonates , Peptides, Cyclic , Micafungin , Bioreactors
2.
Angew Chem Int Ed Engl ; 62(12): e202215529, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36704842

ABSTRACT

Flavonoids are important plant natural products with variable structures and bioactivities. All known plant flavonoids are generated under the catalysis of a type III polyketide synthase (PKS) followed by a chalcone isomerase (CHI) and a flavone synthase (FNS). In this study, the biosynthetic gene cluster of chlorflavonin, a fungal flavonoid with acetolactate synthase inhibitory activity, was discovered using a self-resistance-gene-directed strategy. A novel flavonoid biosynthetic pathway in fungi was revealed. A core nonribosomal peptide synthetase-polyketide synthase (NRPS-PKS) is responsible for the generation of the key precursor chalcone. Then, a new type of CHI catalyzes the conversion of a chalcone into a flavanone by a histidine-mediated oxa-Michael addition mechanism. Finally, the desaturation of flavanone to flavone is catalyzed by a new type of FNS, a flavin mononucleotide (FMN)-dependent oxidoreductase.


Subject(s)
Chalcones , Flavanones , Flavones , Polyketide Synthases/metabolism , Fungi/metabolism , Peptide Synthases/metabolism
3.
Metab Eng ; 74: 160-167, 2022 11.
Article in English | MEDLINE | ID: mdl-36328296

ABSTRACT

Micafungin, a semisynthetic derivative of the cyclic hexapeptide FR901379 produced by Coleophoma empetri fermentation, is the only O-sulfonated echinocandin-type antifungal drug. However, the detailed formation mechanism of O-sulfonate group, whether before or after the assembly of hexapeptide, remains elusive. Here, we confirmed that O-sulfonylation occurs after hexapeptide assembly as a kind of postmodification in the biosynthesis of FR901379. The released cyclic hexapeptide was hydroxylated by cytochrome P450 McfP and successively sulfonated by sulfotransferase McfS. And other three echinocandin sulfotransferases were identified through genome mining by using McfS as a sequence probe. Moreover, pneumocandin B0, the precursor of caspofungin, could be O-sulfonated by heterologously introducing the McfP-McfS into the pneumocandin B0-producing species Glarea lozoyensis. The water-solubility of sulfonated pneumocandin B0 is 4000 times higher than that of pneumocandin B0. The revealed O-sulfonation mechanism will provide new insights into the design and production of novel sulfonated echinocandins by metabolic engineering.


Subject(s)
Antifungal Agents , Echinocandins , Antifungal Agents/metabolism , Echinocandins/metabolism , Fermentation , Metabolic Engineering
4.
Mar Drugs ; 20(8)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36005526

ABSTRACT

Filamentous fungi are abundant resources of bioactive natural products. Here, 151 marine-derived fungi were collected from the north Yellow Sea and identified by an internal transcribed spacer (ITS) sequence. The crude extracts of all strains were evaluated for their antimicrobial activities and analyzed by HPLC fingerprint. Based on these, strain Penicillium oxalicum MEFC104 was selected for further investigation. Two new polyketide-amino acid hybrid compounds with feature structures of tetramic acid, oxopyrrolidine A and B, were isolated. Their planner structures were assigned by HRESIMS and 1D/2D NMR experiments. The absolute configurations were determined by modified Mosher's method, J-based configuration analysis, and ECD calculations. Furthermore, the biosynthetic pathway was identified by bioinformatic analysis and gene-deletion experiments. This study established a link between oxopyrrolidines and the corresponding biosynthesis genes in P. oxalicum.


Subject(s)
Penicillium , Polyketides , Fungi , Penicillium/chemistry , Penicillium/genetics
5.
Fungal Genet Biol ; 160: 103690, 2022 05.
Article in English | MEDLINE | ID: mdl-35351612

ABSTRACT

In filamentous fungi, the secondary metabolism is environmentally sensitive. Most of the biosynthetic gene clusters of secondary metabolites are silent under laboratory conditions. In this study, a highly conserved naphthopyrone PKS ATEG_06206 was identified in the genome sequence of A. terreus MEFC01. This gene is silent under laboratory conditions. To study the function of this PKS, we activated the silent biosynthetic pathway by replacing the promoter of cluster-specific transcriptional factor ATEG_06205 in A. terreus MEFC01. With this strategy, we confirmed that the products of this cryptic PKS are naphthoquinones. These naphthoquinones are soluble pigments, which could be secreted into the agar medium and culture broth. For this reason, the colour of mycelium and conidia of the activated mutant was significantly darker than that of the parental strain. The gene cluster and biosynthetic pathway were further elucidated through the reverse genetics approach and enzymatic assay in vitro.


Subject(s)
Naphthoquinones , Polyketides , Aspergillus , Multigene Family , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Polyketides/metabolism , Secondary Metabolism/genetics
6.
Front Microbiol ; 12: 734780, 2021.
Article in English | MEDLINE | ID: mdl-34489920

ABSTRACT

Micafungin is an important echinocandin antifungal agent for the treatment of invasive fungal infections. In industry, micafungin is derived from the natural product FR901379, which is a non-ribosomal cyclic hexapeptide produced by the filamentous fungus Coleophoma empetri. The difficulty of genetic manipulation in C. empetri restricts the clarification of FR901379 biosynthetic mechanism. In this work, we developed an efficient genetic manipulation system in the industrial FR901379-producing strain C. empetri MEFC009. Firstly, a convenient protoplast-mediated transformation (PMT) method was developed. Secondly, with this transformation method, the essential genetic elements were verified. Selectable markers hph, neo, and nat can be used for the transformation, and promotors Ppgk, PgpdA, and PgpdAt are functional in C. empetri MEFC009. Thirdly, the frequency of homologous recombination was improved from 4 to 100% by deleting the ku80 gene, resulting in an excellent chassis cell for gene-targeting. Additionally, the advantage of this genetic manipulation system was demonstrated in the identification of the polyketide synthase (PKS) responsible for the biosynthesis of dihydroxynapthalene (DHN)-melanin. This genetic manipulation system will be a useful platform for the research of FR901379 and further genome mining of secondary metabolites in C. empetri.

7.
Curr Opin Biotechnol ; 69: 273-280, 2021 06.
Article in English | MEDLINE | ID: mdl-33713917

ABSTRACT

Aspergillus terreus is an important Aspergillus species, which has been applied in the industrial production of the bio-based chemical itaconic acid and the lipid-lowering drug lovastatin. The excellent fermentation capability has been demonstrated in these industrial applications. The genomic information revealed that the outstanding capacity of natural product synthesis by A. terreus remains to be further explored. With advances of the genome mining strategy, the products of several cryptic biosynthetic gene clusters have been discovered recently. In addition, a series of metabolic engineering studies have been performed in the industrial strains of lovastatin and itaconic acid to further improve the production processes. This review presents the current progress and the future outlook in the field of A. terreus biotechnology.


Subject(s)
Aspergillus , Pharmaceutical Preparations , Aspergillus/genetics , Biotechnology , Fermentation , Fungi
8.
Proc Natl Acad Sci U S A ; 116(27): 13305-13310, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209052

ABSTRACT

Mycophenolic acid (MPA) from filamentous fungi is the first natural product antibiotic to be isolated and crystallized, and a first-line immunosuppressive drug for organ transplantations and autoimmune diseases. However, some key biosynthetic mechanisms of such an old and important molecule have remained unclear. Here, we elucidate the MPA biosynthetic pathway that features both compartmentalized enzymatic steps and unique cooperation between biosynthetic and ß-oxidation catabolism machineries based on targeted gene inactivation, feeding experiments in heterologous expression hosts, enzyme functional characterization and kinetic analysis, and microscopic observation of protein subcellular localization. Besides identification of the oxygenase MpaB' as the long-sought key enzyme responsible for the oxidative cleavage of the farnesyl side chain, we reveal the intriguing pattern of compartmentalization for the MPA biosynthetic enzymes, including the cytosolic polyketide synthase MpaC' and O-methyltransferase MpaG', the Golgi apparatus-associated prenyltransferase MpaA', the endoplasmic reticulum-bound oxygenase MpaB' and P450-hydrolase fusion enzyme MpaDE', and the peroxisomal acyl-coenzyme A (CoA) hydrolase MpaH'. The whole pathway is elegantly comediated by these compartmentalized enzymes, together with the peroxisomal ß-oxidation machinery. Beyond characterizing the remaining outstanding steps of the MPA biosynthetic steps, our study highlights the importance of considering subcellular contexts and the broader cellular metabolism in natural product biosynthesis.


Subject(s)
Mycophenolic Acid/metabolism , Aspergillus oryzae/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Metabolic Networks and Pathways , Oxidation-Reduction , Penicillium/metabolism , Peroxisomes/metabolism , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism
9.
J Biol Chem ; 292(17): 7095-7104, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28292933

ABSTRACT

Bafilomycins are an important subgroup of polyketides with diverse biological activities and possible applications as specific inhibitors of vacuolar H+-ATPase. However, the general toxicity and structural complexity of bafilomycins present formidable challenges to drug design via chemical modification, prompting interests in improving bafilomycin activities via biosynthetic approaches. Two bafilomycin biosynthetic gene clusters have been identified, but their post-polyketide synthase (PKS) tailoring steps for structural diversification and bioactivity improvement remain largely unknown. In this study, the post-PKS tailoring pathway from bafilomycin A1 (1)→C1 (2)→B1 (3) in the marine microorganism Streptomyces lohii was elucidated for the first time by in vivo gene inactivation and in vitro biochemical characterization. We found that fumarate is first adenylated by a novel fumarate adenylyltransferase Orf3. Then, the fumaryl transferase Orf2 is responsible for transferring the fumarate moiety from fumaryl-AMP to the 21-hydroxyl group of 1 to generate 2. Last, the ATP-dependent amide synthetase BafY catalyzes the condensation of 2 and 2-amino-3-hydroxycyclopent-2-enone (C5N) produced by the 5-aminolevulinic acid synthase BafZ and the acyl-CoA ligase BafX, giving rise to the final product 3. The elucidation of fumarate incorporation mechanism represents the first paradigm for biosynthesis of natural products containing the fumarate moiety. Moreover, the bafilomycin post-PKS tailoring pathway features an interesting cross-talk between primary and secondary metabolisms for natural product biosynthesis. Taken together, this work provides significant insights into bafilomycin biosynthesis to inform future pharmacological development of these compounds.


Subject(s)
Macrolides/chemistry , Streptomyces/metabolism , 5-Aminolevulinate Synthetase/metabolism , Adenosine Triphosphate/chemistry , Catalysis , Chromatography, High Pressure Liquid , Drug Design , Fumarates/chemistry , Genes, Bacterial , Genetic Vectors , Kinetics , Multigene Family , Open Reading Frames , Polyketide Synthases/metabolism , Polyketides/chemistry , Polymerase Chain Reaction , Vacuolar Proton-Translocating ATPases/metabolism
11.
CNS Neurol Disord Drug Targets ; 11(1): 81-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22229318

ABSTRACT

The pathological lesions typical of Alzheimer disease (AD) are sites of significant and abnormal metal accumulation. Metal chelation therapy, therefore, provides a very attractive therapeutic measure for the neuronal deterioration of AD, though its institution suffers fundamental deficiencies. Namely, chelating agents, which bind to and remove excess transition metals from the body, must penetrate the blood-brain barrier to instill any real effect on the oxidative damages caused by the presence of the metals in the brain. Despite many advances in chelation administration, however, this vital requirement remains therapeutically out of reach: the most effective chelators-i.e., those that have high affinity and specificity for transition metals like iron and copper-are bulky and hydrophilic, making it difficult to reach their physiological place of action. Moreover, small, lipophilic chelators, which can pass through the brain's defensive wall, essentially suffer from their over-effectiveness. That is, they induce toxicity on proliferating cells by removing transition metals from vital RNA enzymes. Fortunately, research has provided a loophole. Nanoparticles, tiny, artificial or natural organic polymers, are capable of transporting metal chelating agents across the blood-brain barrier regardless of their size and hydrophilicity. The compounds can thereby sufficiently ameliorate the oxidative toxicity of excess metals in an AD brain without inducing any such toxicity themselves. We here discuss the current status of nanoparticle delivery systems as they relate to AD chelation therapy and elaborate on their mechanism of action. An exciting future for AD treatment lies ahead.


Subject(s)
Brain/drug effects , Chelating Agents/administration & dosage , Chelation Therapy , Drug Delivery Systems , Nanoparticles/chemistry , Oxidative Stress/drug effects , Transition Elements , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Chelating Agents/pharmacokinetics , Humans , Permeability/drug effects , Receptors, LDL/metabolism
12.
Hemoglobin ; 35(3): 291-300, 2011.
Article in English | MEDLINE | ID: mdl-21599441

ABSTRACT

The only proposed treatment for uranium (U) internal decontamination is chelation therapy, and so far, there is no effective chelating drug available for this purpose. A modified iron chelator of deferiprone (L1) has been investigated for its chemical and pharmacological properties as a new U chelating agent. The results have demonstrated that the chelator, 1-(2-hydroxyethoxy)methyl-2-methyl-3-hydroxyl-4-pyridinone (HEML1), has the ability to chelate uranyl ion, forming a stable complex with 2:1 (chelator/U) stoichiometry. The chelator, HEML1, does not pose any adverse effects on cultured human kidney cells, and shows the capability of protecting the cells against U-associated cytotoxicity and hydrogen peroxide-induced free radical damage. Moreover, because of the hydrophilic property of both HEML1 and its U-complex, the chelator has the potential to target the kidneys where the U prefer to deposit, and to leave the organ after complexing U. These results suggest that HEML1 may be able to serve as a bifunctional therapeutic for internal radionuclide decontamination.


Subject(s)
Antidotes/chemistry , Chelation Therapy/methods , Decontamination/methods , Kidney/pathology , Pyridones/therapeutic use , Uranium/toxicity , Cells, Cultured , Free Radicals , Humans , Hydrogen Peroxide , Kidney/drug effects , Protective Agents/chemistry , Pyridones/pharmacology
13.
Methods Mol Biol ; 610: 123-44, 2010.
Article in English | MEDLINE | ID: mdl-20013176

ABSTRACT

Current therapies for Alzheimer disease (AD) such as the acetylcholinesterase inhibitors and the latest NMDA receptor inhibitor, Namenda, provide moderate symptomatic delay at various stages of the disease, but do not arrest the disease progression or bring in meaningful remission. New approaches to the disease management are urgently needed. Although the etiology of AD is largely unknown, oxidative damage mediated by metals is likely a significant contributor since metals such as iron, aluminum, zinc, and copper are dysregulated and/or increased in AD brain tissue and create a pro-oxidative environment. This role of metal ion-induced free radical formation in AD makes chelation therapy an attractive means of dampening the oxidative stress burden in neurons. The chelator desferrioxamine, FDA approved for iron overload, has shown some benefit in AD, but like many chelators, it has a host of adverse effects and substantial obstacles for tissue-specific targeting. Other chelators are under development and have shown various strengths and weaknesses. Here, we propose a novel system of chelation therapy through the use of nanoparticles. Nanoparticles conjugated to chelators show unique ability to cross the blood-brain barrier (BBB), chelate metals, and exit through the BBB with their corresponding complexed metal ions. This method may provide a safer and more effective means of reducing the metal load in neural tissue, thus attenuating the harmful effects of oxidative damage and its sequelae. Experimental procedures are presented in this chapter.


Subject(s)
Alzheimer Disease/drug therapy , Chelation Therapy/methods , Iron Chelating Agents/therapeutic use , Nanoparticles , Aged , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Benzoates/chemistry , Benzoates/pharmacology , Benzoates/therapeutic use , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Deferasirox , Deferoxamine/chemistry , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Carriers/therapeutic use , Ferritins/chemistry , Humans , Iron/chemistry , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Metals/chemistry , Metals/metabolism , Molecular Structure , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/therapeutic use
14.
J Nanoneurosci ; 1(1): 42-55, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19936278

ABSTRACT

Alzheimer's disease (AD) is a devastating neuro-degenerative disorder characterized by the progressive and irreversible loss of memory followed by complete dementia. Despite the disease's high prevalence and great economic and social burden, an explicative etiology or viable cure is not available. Great effort has been made to better understand the disease's pathogenesis, and to develop more effective therapeutic agents. However, success is greatly hampered by the presence of the blood-brain barrier that limits a large number of potential therapeutics from entering the brain. Nanoparticle-mediated drug delivery is one of the few valuable tools for overcoming this impediment and its application as a potential AD treatment shows promise. In this review, the current studies on nanoparticle delivery of chelation agents as possible therapeutics for AD are discussed because several metals are found excessive in the AD brain and may play a role in the disease development. Specifically, a novel approach involving transport of iron chelation agents into and out of the brain by nanoparticles is highlighted. This approach may provide a safer and more effective means of simultaneously reducing several toxic metals in the AD brain. It may also provide insights into the mechanisms of AD pathophysiology, and prove useful in treating other iron-associated neurodegenerative diseases such as Friedreich's ataxia, Parkinson's disease, Huntington's disease and Hallervorden-Spatz Syndrome. It is important to note that the use of nanoparticle-mediated transport to facilitate toxicant excretion from diseased sites in the body may advance nanoparticle technology, which is currently focused on targeted drug delivery for disease prevention and treatment. The application of nanoparticle-mediated drug transport in the treatment of AD is at its very early stages of development and, therefore, more studies are warranted.

15.
Neurosci Lett ; 455(3): 187-90, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19429118

ABSTRACT

Oxidative stress and amyloid-beta are considered major etiological and pathological factors in the initiation and promotion of neurodegeneration in Alzheimer disease (AD). Insomuch as causes of such oxidative stress, transition metals, such as iron and copper, which are found in high concentrations in the brains of AD patients and accumulate specifically in the pathological lesions, are viewed as key contributors to the altered redox state. Likewise, the aggregation and toxicity of amyloid-beta is dependent upon transition metals. As such, chelating agents that selectively bind to and remove and/or "redox silence" transition metals have long been considered as attractive therapies for AD. However, the blood-brain barrier and neurotoxicity of many traditional metal chelators has limited their utility in AD or other neurodegenerative disorders. To circumvent this, we previously suggested that nanoparticles conjugated to iron chelators may have the potential to deliver chelators into the brain and overcome such issues as chelator bioavailability and toxic side-effects. In this study, we synthesized a prototype nanoparticle-chelator conjugate (Nano-N2PY) and demonstrated its ability to protect human cortical neurons from amyloid-beta-associated oxidative toxicity. Furthermore, Nano-N2PY nanoparticle-chelator conjugates effectively inhibited amyloid-beta aggregate formation. Overall, this study indicates that Nano-N2PY, or other nanoparticles conjugated to metal chelators, may provide a novel therapeutic strategy for AD and other neurodegenerative diseases associated with excess transition metals.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Iron Chelating Agents/therapeutic use , Iron Metabolism Disorders/drug therapy , Nanoparticles/therapeutic use , Nerve Degeneration/drug therapy , Pyridones/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cells, Cultured , Humans , Iron/antagonists & inhibitors , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron Metabolism Disorders/complications , Iron Metabolism Disorders/metabolism , Nanoparticles/chemistry , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Neurotoxins/antagonists & inhibitors , Neurotoxins/metabolism , Plaque, Amyloid/drug effects , Plaque, Amyloid/metabolism , Pyridones/pharmacology
16.
Prog Brain Res ; 180: 97-108, 2009.
Article in English | MEDLINE | ID: mdl-20302830

ABSTRACT

Oxidative stress is known to play a key role in the initiation and promotion of the neurodegeneration that characterizes the pathogenesis of Alzheimer disease (AD). An accumulation of redox active transition metals, including iron and copper, is likely a major generator of reactive oxidative species and other free radicals and is thought to induce a detrimental cycle of oxidative stress, amyloid-beta aggregation, and neurodegeneration. As such, metal chelators may provide an alternative therapeutic approach to sequester redox active metals and prevent the onslaught of oxidative damage. Unfortunately, however, metal chelation approaches are currently limited in their potential, since many cannot readily pass the blood-brain barrier (BBB), due to their hydrophilicity, and many are neurotoxic at high concentrations. To circumvent such issues, here we describe the development of iron chelator-nanoparticle conjugation that allows delivery of target chelator to the brain in the absence of neurotoxicity. Such nanoparticle delivery of iron chelators will likely provide a highly advantageous mode of attack on the oxidative stress that plagues AD as well as other conditions characterized by excess metal accumulation.


Subject(s)
Alzheimer Disease/drug therapy , Drug Delivery Systems/methods , Iron Chelating Agents/chemical synthesis , Nanoparticles/chemistry , Nanotechnology/methods , Neuropharmacology/methods , Animals , Dimerization , Drug Delivery Systems/trends , Drug Design , Humans , Iron Chelating Agents/pharmacokinetics , Nanoparticles/therapeutic use , Nanotechnology/trends , Neuropharmacology/trends , Oxidative Stress/drug effects , Oxidative Stress/physiology
17.
Hemoglobin ; 32(1-2): 181-90, 2008.
Article in English | MEDLINE | ID: mdl-18274995

ABSTRACT

Earlier studies revealed that age-associated iron accumulation plays an important causal role in osteopenic development after estrogen deficiency. It is believed that an increase in iron content is associated with an increased likelihood of oxidative damage at the point of iron accumulation. However, there is no direct evidence that the iron accumulated in skeletal tissue causes free radical oxidative damage and consequent bone loss. Iron depletion from skeletal tissues of ovariectomized (OVX) rats was carried out with the oral chelator [1-N-docosyl-triethylenetetraminepentaacetic acid (DoTTPA)]. Results suggest the causal role of iron in oxidative damage that may lead to bone loss in the rats. The results also show the therapeutic potential of the bone-targeted chelator to protect against bone loss associated with age-iron accumulation as well as iron overload diseases.


Subject(s)
Acetates/therapeutic use , Iron Chelating Agents/therapeutic use , Iron/metabolism , Muscle, Skeletal/metabolism , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/prevention & control , Trientine/analogs & derivatives , Acetates/administration & dosage , Acetates/chemistry , Acetates/metabolism , Animals , Electron Spin Resonance Spectroscopy , Female , Free Radicals/metabolism , Humans , Iron Chelating Agents/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Trientine/administration & dosage , Trientine/chemistry , Trientine/metabolism , Trientine/therapeutic use
18.
Neurosci Lett ; 406(3): 189-93, 2006 Oct 09.
Article in English | MEDLINE | ID: mdl-16919875

ABSTRACT

Accumulating evidence suggests that oxidative stress may be a major etiologic factor in initiating and promoting neurodegeneration in Alzheimer disease. Contributing to this, there is a dyshomeostasis of metal ions in Alzheimer disease with abnormally high levels of redox-active metals, particularly iron, in affected areas of the brain. Although it is unclear whether metal excesses are the sole cause of oxidative stress and neurodegeneration or a by-product of neuronal loss, the finding that metal chelators can partially solubilize amyloid-beta deposits in Alzheimer disease suggests a promising therapeutic role for chelating agents. However, the blood-brain barrier and toxicity of known chelators limit their utility. In this study, we suggest that covalent conjugation of iron chelators with nanoparticles may help overcome the limitations in blood-brain barrier permeability of existing chelation therapy. Using in vitro studies, we have shown that a chelator-nanoparticle system and the chelator-nanoparticle system complexed with iron, when incubated with human plasma, preferentially adsorb apolipoprotein E and apolipoprotein A-I, that would facilitate transport into and out of the brain via mechanisms used for transporting low-density lipoprotein. Our studies suggest a unique approach, utilizing nanoparticles, to transport chelators and chelator-metal complexes in both directions across the blood-brain barrier, thus providing safer and more effective chelation treatment in Alzheimer disease and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/pathology , Brain/drug effects , Iron Chelating Agents/pharmacology , Iron/metabolism , Nanostructures , Pyridones/pharmacology , Aged, 80 and over , Apolipoprotein A-I/metabolism , Apolipoproteins E/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Humans , In Vitro Techniques , Iron Chelating Agents/chemistry , Pyridones/chemistry
19.
Biometals ; 19(3): 245-51, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16691320

ABSTRACT

Iron accumulation in tissues is believed to be a characteristic of aged humans and a risk factor for some chronic diseases. However, it is not known whether age-associated iron accumulation is part of the pathogenesis of postmenopausal osteoporosis that affects approximately one out three women worldwide. Here, we confirmed that this accumulation of iron was associated with osteopenia in ovariectomized (OVX) rats (a model of peri- and postmenopausal osteoporosis due to estrogen deficiency). To further investigate whether the increased iron level plays a causal role in the onset of bone loss, we treated OVX rats with an orally active and bone targeted chelator that prevented iron accumulation in their skeletal tissues. The results showed that this treatment mitigated the loss of bone mass and the deterioration of bone micro-architecture. We also found that one possible mechanism of the protective action of iron chelation was to significantly reduce bone resorption. Thus, these findings provide a novel target and a potentially useful therapeutic strategy for the prevention and treatment of postmenopausal osteoporosis and perhaps other age-related diseases.


Subject(s)
Acetates/therapeutic use , Aging/metabolism , Iron Chelating Agents/therapeutic use , Iron/metabolism , Osteoporosis, Postmenopausal/physiopathology , Trientine/analogs & derivatives , Animals , Bone Density/drug effects , Chelation Therapy , Female , Humans , Osteoporosis, Postmenopausal/drug therapy , Osteoporosis, Postmenopausal/prevention & control , Ovariectomy , Ovary/physiology , Rats , Rats, Sprague-Dawley , Trientine/therapeutic use
20.
Biochim Biophys Acta ; 1741(3): 246-52, 2005 Sep 25.
Article in English | MEDLINE | ID: mdl-16051470

ABSTRACT

Current therapies for Alzheimer disease (AD) such as the anticholinesterase inhibitors and the latest NMDA receptor inhibitor, Namenda, provide moderate symptomatic delay at various stages of disease, but do not arrest disease progression or supply meaningful remission. As such, new approaches to disease management are urgently needed. Although the etiology of AD is largely unknown, oxidative damage mediated by metals is likely a significant contributor since metals such as iron, aluminum, zinc, and copper are dysregulated and/or increased in AD brain tissue and create a pro-oxidative environment. This role of metal ion-induced free radical formation in AD makes chelation therapy an attractive means of dampening the oxidative stress burden in neurons. The chelator desferioxamine, FDA approved for iron overload, has shown some benefit in AD, but like many chelators, it has a host of adverse effects and substantial obstacles for tissue-specific targeting. Other chelators are under development and have shown various strengths and weaknesses. In this review, we propose a novel system of chelation therapy through the use of nanoparticles. Nanoparticles conjugated to chelators show a unique ability to cross the blood-brain barrier (BBB), chelate metals, and exit through the BBB with their corresponding complexed metal ions. This method may prove to be a safe and effective means of reducing the metal load in neural tissue thus staving off the harmful effects of oxidative damage and its sequelae.


Subject(s)
Alzheimer Disease/drug therapy , Chelating Agents/therapeutic use , Chelation Therapy/methods , Nanostructures , Blood-Brain Barrier/metabolism , Chelating Agents/metabolism , Humans , Permeability
SELECTION OF CITATIONS
SEARCH DETAIL
...